Next Article in Journal
Medicinal Plants against Viral Infections: A Review of Metabolomics Evidence for the Antiviral Properties and Potentials in Plant Sources
Next Article in Special Issue
Mechanisms and Cardiorenal Complications of Chronic Anemia in People with HIV
Previous Article in Journal
The Potential of Usnic-Acid-Based Thiazolo-Thiophenes as Inhibitors of the Main Protease of SARS-CoV-2 Viruses
Previous Article in Special Issue
Dynamics of Low-Level Viremia and Immune Activation after Switching to a Darunavir-Based Regimen
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Examining Chronic Inflammation, Immune Metabolism, and T Cell Dysfunction in HIV Infection

by
Wenli Mu
1,2,
Vaibhavi Patankar
1,2,
Scott Kitchen
1,2 and
Anjie Zhen
1,2,*
1
Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
2
UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
*
Author to whom correspondence should be addressed.
Viruses 2024, 16(2), 219; https://doi.org/10.3390/v16020219
Submission received: 28 November 2023 / Revised: 26 January 2024 / Accepted: 29 January 2024 / Published: 31 January 2024

Abstract

:
Chronic Human Immunodeficiency Virus (HIV) infection remains a significant challenge to global public health. Despite advances in antiretroviral therapy (ART), which has transformed HIV infection from a fatal disease into a manageable chronic condition, a definitive cure remains elusive. One of the key features of HIV infection is chronic immune activation and inflammation, which are strongly associated with, and predictive of, HIV disease progression, even in patients successfully treated with suppressive ART. Chronic inflammation is characterized by persistent inflammation, immune cell metabolic dysregulation, and cellular exhaustion and dysfunction. This review aims to summarize current knowledge of the interplay between chronic inflammation, immune metabolism, and T cell dysfunction in HIV infection, and also discusses the use of humanized mice models to study HIV immune pathogenesis and develop novel therapeutic strategies.

1. Introduction

Chronic human immunodeficiency virus (HIV) infection continues to pose a formidable threat to global health. HIV primarily infects CD4+ T cells, which are crucial for defending the body against various infections and diseases. As the disease progresses in people living with HIV (PLWH), HIV infection is, when compared to uninfected individuals [1,2,3], seen to drive the persistence of higher levels of immune activation and inflammation, which are both a hallmark of the body’s continuous battle against the virus. Despite the success of antiretroviral therapy in controlling viral replication [4,5], there is a growing appreciation that many PLWH, despite successful ART, continue to exhibit signs of chronic, low-grade inflammation [6,7], which is believed to contribute to a range of non-AIDS-related co-morbidities [8,9,10,11,12,13,14]. One of the consequences of chronic inflammation is T cell exhaustion, in which these critical immune cells become less effective, with diminishing capacity to eliminate infected cells effectively [15,16,17]. In addition, the imbalance of metabolic processes, whether directly caused by HIV infection or indirectly by HIV-driven inflammatory responses within immune cells, further contributes to immune activation and dysfunction (summarized in Figure 1) [18,19,20,21]. The combined impact of inflammation, metabolic alterations, and T cell exhaustion underscores the complexities inherent in managing HIV and other HIV-associated disorders [6,22,23]. In this review, we focus on the current understanding of chronic inflammation, immune metabolism, and T cell exhaustion in the wider context of HIV infection in PLWH. We summarize various model system used in HIV research and emphasize the unique advantages of using humanized mice to understand HIV pathogenesis, by using humanized mouse models to investigate the pathogenesis of HIV infection, and especially its relation to immune activation, metabolism, and T cell dysfunction in these novel experimental systems. Understanding these processes is crucial for the development of novel therapeutic strategies that improve the health outcomes of PLWH.

2. Chronic Inflammation and Immune Activation Are Hallmarks of HIV Infection

Persistent inflammation in PLWH is characterized by the continuous activation of various immune cells, including T cells [24,25,26,27], B cells [28,29,30], and monocytes [11,31,32,33]; and elevated levels of pro-inflammatory cytokines [34,35,36], including tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and C-reactive protein (CRP), which contribute to increased morbidity and mortality in PLWH [3,34,37]. Several factors contribute to immune activation/inflammation during chronic HIV infection, including persistent viral replication, microbial translocation, and co-infections with other pathogens [3]. Despite effective, highly suppressive ART, latently infected cells can reactivate and produce new virions, contributing to persistent viral replication [38,39,40]. Certain anatomical locations, such as the lymph nodes [41] and the central nervous system [42], serve as persistent reservoirs, due to limited ART access and ineffective immune surveillance [43], which leads to continuous activation of the immune system and production of inflammatory mediators [44,45]. Additionally, the products of HIV expression induce inflammation by activating various signaling pathways. For example, the HIV protein gp120 has been shown to activate the NF-κB pathway, leading to the production of proinflammatory cytokines [46]. Residual viral particles, as well as Tat and Nef proteins, can also induce cellular activation and the production of inflammatory cytokines [47,48,49,50].
Type I interferons (IFN-Is), including IFN-α and IFN-β, are central components of the innate immune response. IFN-Is are rapidly induced by viral infection through pattern recognition receptors (PRRs) and intracellular proteins that recognize direct cellular infection [51]. HIV infection is rapidly sensed by PRRs and cytosolic sensors that detect viral cDNA or RNAs, which leads to the production of IFN-Is and the expression of IFN-simulated genes (ISGs), which are key effector molecules that exhibit anti-viral activities [52,53,54]. Type I interferon is also critical for the induction of functionally optimal antigen- specific CD8 T cells in HIV infection [55]. However, HIV interferes with the IFN-I responses by impairing functions of ISGs and viral isolates shown to have heightened IFN-I resistance at transmission and ART interruption [56,57]. In addition to their antiviral functions, particularly during acute infections, IFN-Is also have critical immune modulating capacities, and are associated with chronic inflammation in many disease states [51,58,59]. IFN-Is can play a dichotomous role and drive an immunosuppressive and exhausted immune state during chronic infection [59]. Elevated IFN-I-stimulated gene (termed ISG) expression is upregulated in HIV infection, remains elevated despite suppressive ART, and is correlated with disease progression [22,60]. As a result, chronic IFN-I signaling has emerged as a prime suspect in the driving of immune activation and HIV disease progression [59,61,62,63]. Animal study research has shown that blocking type I interferon signaling during chronic infection leads to the restoration of T cell functions and a reduced reservoir [62,64,65]. Additional studies are needed to evaluate if IFN blockade can act as a supplement to ART and improve immune function [66].
Microbial translocation is another major contributor to chronic inflammation [67,68]. Damage to the gut mucosal barrier during acute HIV infection allows the translocation of microbial products, such as lipopolysaccharide (LPS), from the gut lumen into the systemic circulation. This microbial translocation further stimulates the immune system and contributes to systemic inflammation [69]. Lastly, coinfections with other pathogens, such as the hepatitis C virus (HCV), cytomegalovirus (CMV), and mycobacterium tuberculosis, are common in PLWH [70,71,72]. These coinfections activate the immune system, exacerbating chronic inflammation and leading to the increased production of inflammatory cytokines and chemokines, which further drives HIV infection and pathogenesis that can also impact the effectiveness of ART [72].

3. Metabolic Stress during HIV Infection

Uncontrolled HIV infection results in progressive CD4 T cell depletion, impairment of both B cell and cytotoxic T cell responses, and ultimately leads to system immune failure and acquired immunodeficiency (AIDS) [73]. Despite the effect of ART, the virus cannot be completely eradicated, and its persistence supports a chronic status of immune activation and immune system dysfunction [22]. As a result, PLWH experience various systemic challenges, including metabolic stress. One of the highly prevalent metabolic dysregulations occurs with lipid metabolism, such as lower levels of high-density lipoprotein (HDL) cholesterol, increased low-density (LDL) lipoprotein, total (TC) cholesterol and triglycerides, leading to dyslipidemia being observed in many PLWH [74,75,76]. Several viral proteins are implicated in dyslipidemia. For example, HIV accessory protein Nef down regulates the adenosine-triphosphate-binding cassette transporter A1 (ABCA1), resulting in reduced efflux of cholesterol to HDL and lipid accumulation in infected macrophages [77]. Moreover, HIV replication is associated with the increase of fatty acid synthase activity, which leads to increased levels of free fatty acids and LDLs [78]. In addition, HIV-mediated immune activation alters lipid processing and transportation, and can lead to production of lipid species that are more ‘inflammatory’, such as oxidized forms of LDL (oxLDL) and HDL (HDLox) [79], forming a vicious cycle of inflammation. Glucose metabolism irregularities, such as insulin resistance [80], which is correlated with coronary artery stenosis [81], are another abnormality associated with HIV infection. Insulin resistance is associated with elevated proinflammatory cytokines and the activation of innate responses, such as toll-like receptors (TLRs), inducible nitric oxide synthase (iNOS), protein kinase R (PKR), c-Jun N-terminal kinase (JNK), and NF-κB, which are connected to insulin receptor and its downstream signaling pathway IRS/PI3k/Akt [82]. Interestingly, a recent study reported that increased monocyte inflammatory responses to oxLDL are associated with insulin resistance in PLWH [83], and noted defects in cholesterol homeostasis and lipid raft impairment are connected to insulin resistance [84,85]. Both findings suggest that factors of metabolic stress are interconnected and exacerbated by systemic inflammation.
Effective ART can, in general, improve the metabolic profile by reducing heightened inflammation and mitigating the inherent effects of HIV replication on metabolism. Nevertheless, patients on ART exhibit significant metabolic stress and antiretroviral drugs can themselves cause metabolic disorders [86]. Classes of ART include nucleoside-analog reverse transcriptase inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), protease inhibitors (PIs), integrase inhibitors (INIs), fusion inhibitors, and coreceptor antagonists, which each interfere with critical steps in the viral replication lifecycle. NRTIs inhibit DNA polymerase gamma (Pol-γ), which functions in mitochondria DNA (mtDNA) replication and maintenance, and have therefore been implicated in mitochondria toxicity [87]. NRTI may be incorporated into mtDNA via Pol-γ by competing with natural thymidine triphosphate, leading to the mutation or termination of mtDNA. In addition, NRTIs have been shown to impair ATP synthesis, increase oxidative stress, and decrease mitochondria membrane potential Ψm [88] and have, as a result, been linked to long-term metabolic and cardiovascular complications, such as mitochondria toxicity, lactic acidosis, and lipodystrophy [89,90,91,92,93]. NNRTIs inhibit viral replication by binding to a hydrophobic pocket adjacent to the active site of HIV reverse transcriptase. Efavirenz (EFV), a common NNRTI, has been shown to increase oxidative stress, decrease Ψm and induce apoptosis [94]. Protease Inhibitors (PIs), another common class of ART, interfere with the cleavage of essential viral maturation polyprotein precursors by inhibiting HIV protease. PIs, such as ritonavir, have been shown to induce oxidative stress, decrease Ψm and ATP production, and inhibit cholesterol efflux, leading to side effects associated with metabolic disturbances, including dyslipidemia, lipodystrophy, and insulin resistance [89,90,91,93]. Mitochondria toxicity is particularly pronounced in older drugs such as didanosine (ddI) and stavudine (d4T), but is less common in the newer drugs such as lamivudine (3TC), emtricitabine (FTC) and tenofovir (TDF) [95,96]. Nonetheless, 3TC, FTC and TDF were still shown to decrease fat mtDNA content and affect complex I and IV activity levels [97]. The mechanisms of metabolic altercation by HIV infection and ART are complex, multifactorial and not fully understood, and further studies are required to improve clinical management and the healthy lifespan of PLWH.
Metabolic stress driven by HIV infection has a direct impact on immune cell functions [98,99]. Serum and plasma derived from PLWH revealed altered metabolites of lipid and fatty acids, which may play an important role in driving immune dysfunction [100,101,102]. HIV infection-mediated chronic inflammation also leads to increased lipolysis and altered lipid trafficking [103,104], which can lead to the accumulation of lipid droplets in immune cells, and in turn have various effects on their function [105,106]. For example, lipid metabolite long-chain fatty acid inhibits IFN-γ production by stimulating intraepithelial lymphocytes [107], and inhibits T-cell responses by increasing mitochondrial reactive oxygen species (ROS) [100]. Lipid metabolism plays a key role in macrophage function and IFN-I antiviral responses [108,109]. The excessive accumulation of lipid in monocytes can lead to macrophage foam cell formation, which produces high levels of proinflammatory cytokines and promotes atherosclerotic plaque formation [110]. In addition, monocytes and macrophages with excessive lipid also display altered type I IFN responses [111]. Lastly, viral infection and the proinflammatory cytokines TNF and IL-1beta can induce mitochondria stress, resulting in the release of mtDNA and activation of cGAS-MITA/STING, which in turn activates IFN-I and inflammasome signaling [112]. Moreover, damaged mitochondria can also release mtRNA, ROS, and other mitochondria damage-associated molecular patterns (mtDAMPs), triggering innate signaling, leading to further exacerbation of chronic immune activation during HIV infection [113].
Growing evidence indicates that cellular metabolism plays a key role in supporting immune cell maintenance and development, and also guides immune activation and differentiation [114]. Due to these metabolic perturbations observed in PLWH, it is therefore critical to study immune metabolism and its role in HIV pathogenesis and immune exhaustion [115].

4. Immune Activation and Metabolic Dysfunction Contribute to T Cell Exhaustion during Chronic HIV Infection

T cell exhaustion is a state of T cell dysfunction characterized by the progressive loss of effector activities, the sustained expression of inhibitory receptors, and metabolic alterations [116]. This phenomenon is observed in cancers and various chronic viral infections, including HIV, and is closely associated with the inability of the immune response to adequately control these conditions [116,117,118]. Exhausted T cells exhibit impaired proliferative ability, cytokine production, and cytotoxic activity, which results in ineffective cellular immune responses [15,116,119]. T-cell exhaustion in chronic viral infections is mainly triggered by the persistent activation of TCR signaling, leading to the increased expression of inhibitory and co-inhibitory receptors, such as PD-1, CTLA-4, TIM-3, 2B4, LAG-3, and CD160 [116,120]. While these molecules have important roles in normal immune functions in acute conditions, their upregulation in chronic conditions, such as HIV infection, are highly associated with immune dysfunction. During chronic HIV infection, the upregulation of inhibitory receptors (or called checkpoint inhibitors) in T cells and engagement with their ligands suppresses T cell activation and function. This persistent inhibitory signaling, combined with altered gene expression patterns, leads to T cell exhaustion and compromised antiviral responses [15,121].
Emerging evidence suggests that metabolic distress also contributes to T cell exhaustion and dysfunction. Healthy immune cells maintain a balanced metabolic state, and primarily rely on oxidative phosphorylation (OXPHOS) in mitochondria for energy production in resting conditions. This metabolic pathway is oxygen-dependent and generates more ATP, compared to glycolysis [114,122,123], and this process supports the basic functions of immune cells without promoting excessive proliferation or activation. When immune cells are activated in response to pathogens or other stimuli, they undergo the “Warburg effect”, a metabolic shift to aerobic glycolysis [124,125]. The intensification of aerobic glycolysis allows cells to rapidly generate energy by converting glucose-derived pyruvate into lactate under normoxic conditions, rather than entering the TCA cycle in mitochondria [126]. This metabolic shift plays a crucial role in supporting biosynthetic demands for the activation and proliferation of T cells [103,127]. Chronic immune activation means that immune cells, including T cells and macrophages, are continuously activated and proliferating, which increases their energy and nutrient demands [124,125]. Interestingly, during acute infection, HIV-1 induces the association of NLRX1 with the mitochondria protein FASKD5 to promote OXPHOS and viral replication in CD4 T cells, and viral load setpoint is positively correlated with the OXPHOS pathway [128]. In contrast, during chronic HIV infection, the metabolic demands can lead to nutrient deprivation and the accumulation of metabolic waste products, which in turn affects T cell functions [21,129]. For example, Loisel-Meyer et al. found that HIV-infected macrophages produce higher levels of lactate, which can accumulate in the tissue microenvironment and contribute to T cell dysfunction [130]. Additionally, ROS production is increased in T cells during HIV infection, contributing to oxidative stress and subsequent T cell dysfunction [131,132]. ART, particularly NRTI-mediated mitochondria toxicity, as described above, can also contribute to decreased mitochondrial OXPHOS activity. Therefore, in chronic HIV infection, OXPHOS is decreased in the peripheral blood monocular cells (PBMCs) of PLWH, and is associated with immune dysregulation [133].
During HIV infection, glucose and glutamine metabolism undergoes significant alteration, in both HIV-infected cells and activated immune cells responding to infection [134,135,136,137]. Compared to uninfected cells, there is increased glucose and glutamine metabolic activity in HIV-infected CD4+ T cells and macrophages [138,139]. Activation of CD4 T cells also leads to increased glucose uptake and the expression of glucose transporters [135,140,141]. Increased glycolytic flux is required for viral production and increases the propensity of CD4 T cells to show apoptosis [130]. As a result, increased glucose metabolic activity and increased Glut1 expression are associated with CD4 T cell activation and depletion during chronic HIV infection in PLWHs, and are not completely normalized by ART [141]. Dysregulation of glutamine metabolism in HIV infection can lead to immune cell dysfunction, as evidenced by a negative correlation between glutamine levels and the production of cytokines and chemokines by CD8+ T cells [142,143]; meanwhile, CD4 cell count is inversely correlated with both glutamine and glucose concentrations [144]. In addition, the metabolism of amino acids such as tryptophan and arginine, which are crucial for immune cell function [145,146], are also impaired during HIV infection. Persistent inflammation and immune activation can lead to the depletion of these amino acids. For example, Indoleamine 2,3-dioxygenase (IDO) is an enzyme that is upregulated during inflammation and degrades tryptophan [147]. Increased IDO activity during HIV infection leads to tryptophan depletion, which can have immunosuppressive effects and contribute to T cell dysfunction [148,149].
Early-stage exhausted T cells exhibit a unique metabolic profile, characterized by reduced glycolysis and increased fatty acid oxidation (FAO); however, these cells exhibited impaired mitochondrial function [21,129]. This metabolic shift from aerobic glycolysis is triggered by continuous antigen exposure, which upregulates the PD-1/PD-L1 pathway, resulting in inhibition of TCR/CD28-mediated PI3K signaling and reduced glycolysis and glutamine utilization in effector T cells [19,150]. In contrast, the terminal stage of exhausted T cells mainly relies on glycolytic metabolism with impaired glycolysis and OXPHOS [151,152,153]. Notably, the decline in glycolysis and the mitochondrial respiration of T cells is observed before the onset of T cell dysfunction in early chronic infection, suggesting that metabolic abnormalities set in before, and not as a result of, T cell exhaustion [19,154]. Therefore, modulating metabolism may provide a feasible and efficient strategy to prevent T cell exhaustion in chronic viral infections.
In summary, immune activation, immune cell metabolic dysfunction and exhaustion are intricately linked in a complex, bidirectional relationship rather than a straightforward cause-and-effect sequence. Immune activation and metabolic dysfunction are direct contributors to T cell exhaustion, while metabolic dysfunction and immune cell dysfunction can further exacerbate immune activation/chronic inflammation. For example, mitochondrial dysfunction can lead to the production of ROS and the release of mitochondrial DNA, which can activate innate immune pathways and contribute to chronic inflammation [113,155]. Understanding the interactions between immune activation, metabolism dysfunction and immune exhaustion during HIV infection is important, and will contribute to research that seeks to generate new therapeutic approaches to HIV infection. Achieving a better understanding of the molecular mechanisms that underlie T cell exhaustion will ultimately help to address current barriers that inhibit the development of more effective therapies. This will however only be achieved by applying various representative models to analyze these interactions.

5. Current Approaches to Modeling Pathogenesis and Studying the Antiviral Immunity of HIV Infection

Exploring the interplay between immune exhaustion and metabolic dysfunction in HIV pathogenesis requires a multidisciplinary approach. Various models have a distinct role to play in studying immune activation and metabolic dysfunction that can lead to T cell exhaustion.
In vitro studies that examine primary cells and cell lines exposed to HIV-1 or HIV-1 proteins have provided crucial insights into the mechanisms that drive immune dysfunction [15,156,157], and are essential for understanding the molecular and signaling pathways that are involved in immune activation [15,158,159] and metabolic stress [134,160]. Ex vivo studies, on the other hand, can bridge the gap between in vitro studies and in vivo clinical observations, offering a controlled environment to study the HIV-induced alterations in T cell function and metabolism that contribute to the disease pathology [161,162,163,164,165]. However, both in vitro and ex vivo studies lack the complexity of living organisms and cannot replicate systemic responses. The ultimate source of direct evidence is clinical trials, which provide invaluable data for advancing our understanding of HIV pathogenesis and anti-viral immunity, and grasping clinical implications [166,167], however, clinical research faces practical and ethical challenges, as well as constraints related to the limited availability of tissue sampling.
Alternatively, animal models provide unique opportunities to explore therapeutic and prevention approaches, advance HIV-1 management, and gain insight into the mechanism of HIV-1 pathogenesis. Current animal models for the study of HIV-1 infection include non-human primates and humanized mice. Primate models, owing to a genetic and physiological similarity to humans, are critical for understanding the systemic aspects of immune activation [168,169,170] and viral pathogenesis [171,172,173,174,175]. They have been crucial for observing the progression of T cell dysfunction and realistically depict immune exhaustion in HIV-1 infection [176,177,178,179,180,181,182]. However, the use of nonhuman primate models is constrained by ethical considerations, high maintenance cost, and the limited availability of suitable species, and these constraints have restricted experimental group sizes and limited the assessment of various conditions and parameters. In addition, interactions specific to HIV and human host cells cannot be fully assessed in nonhuman primates, since they are typically infected with simian immunodeficiency virus (SIV) or simian-human immunodeficiency virus (SHIV) [183,184,185,186]. Humanized mice models offer a complementary approach by addressing these limitations and opening up the possibility of investigating interactions specific to HIV-1.
Humanized mice are immunodeficient mice engrafted with human cells and/or tissue that have become increasingly valuable as small animal models, both for the close examination of various human diseases and the development of therapeutic strategies [187,188,189].
When compared to primate models of SIV or SHIV, as well as human clinical trials, humanized mouse models of HIV infection are found to possess all the benefits of small animal models: they raise fewer ethical concerns; are less costly; recapitulate in vivo complexity; permit sampling and intervention that are not feasible in clinical trials; and allow a larger sample size, enabling statistically robust studies, which may not be feasible with primate models. In particular, humanized mouse models reconstituted with human immune cells have achieved significant breakthroughs in improved immune reconstitution and have, in recent years, been increasingly widely used in studies of human immunology, infectious diseases, and tumor therapies [187,188,189]. Humanized mice reconstituted with human T cells and other immune cells can support robust HIV infection and HIV latency; have been widely used to study the biology of HIV infection, pathogenesis and anti-HIV immunity; and have also played a critical role in the testing and development of ART and gene- and cell-based therapeutics [190,191,192,193]. Importantly, the model allows the examination of novel therapies that involve the manipulation of human genetics (such as CCR5 knockdown/gene editing), human cell-based immunotherapy (such as CAR-T cell and NK cell therapy), and human biologics (such as the anti-HIV broad neutralizing antibody (bNAb), checkpoint inhibitor therapy, and cytokine treatment, etc.). As a result, the humanized mouse model has emerged as a popular pre-clinical model. Although the murine drug metabolism is different from the human counterpart, the humanized mouse model still provides a versatile model that can be used to explore immune metabolism, and this is because it more closely approximates to human responses than traditional mouse models.
There are many various types of humanized mouse models, which primarily differ in the background mouse strain and humanization procedure. The humanized mouse models most frequently used for HIV research include:
  • Hu-PBL-SCID mice: This model involves transplanting severe combined immunodeficient (SCID) mice with human periphery blood mononuclear cells (PBMCs) [194]. The hu-PBL-SCID models are susceptible to rapid, potent HIV infection and are therefore good models for studying CD4+ T cell depletion and testing anti-viral compounds [189]. However, the hu-PBL-SCID model’s high susceptibility to developing Graft versus host disease (GVHD) in a relatively short period of time makes it a less than ideal candidate for long-term studies [195].
  • Hu-CD34 mice: hu-CD34 mice are generated by engrafting human CD34+ hematopoietic stem/progenitor cells (HSPCs), isolated from adult bone marrow tissue, adult mobilized peripheral blood, umbilical cord blood, or fetal liver, into immunodeficient mice, such as NOD-PrkdcscidIl2rgtmiwjl/Sz (NSG) mice. These mice can support the establishment of a robust human immune system (consisting of T cells, B cells, and myeloid cells, with limited GVHD), and are capable of modeling HIV replication in vivo [189,194]. Although this model supports sustained HIV infection and the establishment of latent/persistently infected cellular reservoirs, the mouse thymus does not support the development of fully functional T cells, resulting in lower levels of T cell reconstitution than the BLT mouse (see below) and non-fully functional T cells. This makes it difficult to study the impact of HIV infection on thymic T cell differentiation and T cell functions [196,197].
  • BLT (humanized bone marrow-liver-thymus) mice: BLT-humanized mice are generated by implanting human fetal liver and thymus tissues into conditioned NSG mice, and simultaneously injecting autologous CD34 HSPCs from a fetal liver [189]. This model allows the development of a robust human immune system, including T cells, B cells, NK cells, and myeloid cells. The humanized BLT mouse model is a powerful small animal model that enables robust human immune reconstitution and robust, natural T cell thymic development, allowing for the comprehensive study of HIV immunity. The model is key to seminal studies of cell and gene therapy and, ultimately, to the discovery of a HIV cure [196,198,199,200,201,202,203,204,205,206,207,208,209,210,211]. It has also contributed to studies of HIV latency [200,212,213,214], and mechanistic studies of HIV immunopathogenesis [64,65,196,215,216,217,218]. Despite its notable advantages, this model presents a number of challenges, including expense, the difficulties of surgical procedures, the procurement of fetal tissues, and the inconsistency between the graft and host disease development [189].
In addition to the aforementioned ways of generating humanized mouse models, the development of new strains of immunodeficient mice has further improved multi-lineage immune reconstitution and the versatility of the humanized mice model [219]. These include but are not limited to, the TKO (C57BL/6 Rag2−/−γc−/−CD47−/−) strain, with deleted CD47 to induce tolerance and reduce GVHD development [220]; MISTRG (C;129S4-Rag2tm1.1Flv Csf1tm1(CSF1)Flv Csf2/Il3tm1.1(CSF2,IL3)Flv Thpotm1.1(TPO)Flv.
Il2rgtm1.1Flv Tg(SIRPA), harbors humanized knock-in alleles M-CSF, IL-3/GM-CSF and TPO, and supports improved innate responses and myeloid differentiation [221]; NSG-SGM3 (NOD-scid IL2Rgnull-3/GM/SF), carries human IL-3, GM-SF and CSF and allows stable myeloid lineage engraftment [222,223,224]; NSG-Tg(hIL34) carries humanized IL-34 and allows the improved engrafting of microglial cells [225]; NSG-Tg (hIL15) carries humanized IL-15 and allows improved Treg and natural killer cell development [226,227]; NSG-A2 expresses human HLA class I A2 molecule supports development of A2 restricted human T cells [228]; and DRAG, which are NOD.Rag1KO.IL2RccKO mice that express HLA-DR4 (0401), shows improved B cell and IgG reconstitution [229].
Each of these models has its own advantages and limitations, and the choice of model depends on the specific research questions being addressed. Among them, hu-CD34 and BLT humanized mice can sustain a chronic HIV infection, which allows researchers to study the long-term interactions between HIV and the human immune system in vivo. Several studies that use humanized mice models have shed light on the mechanisms underlying chronic inflammation and immune exhaustion during HIV infection, with particular emphasis on type I interferon signaling, checkpoint inhibitor expression, inflammasome activation, and cellular metabolic processes. By using the humanized NSG-BLT mouse model, we [65], and others [61,64,230], showed that the chronic immune activation and T cell dysfunction seen in BLT mice after HIV infection resemble the patterns observed in HIV+ patients [231,232]. Importantly, we and others [61,64,65,230] also showed that blocking persistent IFN-I signaling in vivo restored dysfunctional anti-HIV specific T cells, lowered viral loads, and reduced the HIV reservoir. Moreover, our recent study demonstrated that modulating type I IFN signaling with autophagy inducer rapamycin in HIV- infected humanized mice led to decreased immune activation, improved anti-HIV T cell function, produced faster viral suppression during ART, and significantly reduced viral rebound after ART withdrawal [233], further suggesting the pathogenic role of type I interferon during chronic HIV infection.
In addition to chronic type I IFN signaling and T cell exhaustion, HIV-infected humanized mice have also been demonstrated to have elevated soluble inflammatory markers [234,235], increased inflammasome activation [236] and high levels of immune check point inhibitor PD-1 expression in T cells [237], reiterating what has already been seen in PLWH. This has enabled numerous studies that closely examine many different aspects of HIV-induced inflammation in vivo. Studies have shown that blocking PD-1 with an anti-PD-1 antibody in HIV-infected humanized mice led to enhanced T cell responses and reduced viral loads [208,238]. Studies investigating the role of inflammasome showed that a caspase 1 inhibitor can mitigate inflammasome activation and CD4 T cell depletion, and reduce viral load in HIV-infected huCD34 humanized mice [236].
Growing evidence indicates that humanized mice can also be used to study immune metabolism and related therapeutics. For example, induced high cholesterol levels contribute to the proliferation of T cells and T cell–mediated inflammatory diseases in BLT humanized mice [239]. Guo et al. have, in studies using human CD4 T cell-reconstituted mice, investigated the role of OXPHOS in HIV infection. The study demonstrated that metformin treatment inhibits OXPHOS, which targets mitochondrial respiratory chain complex-I, and suppresses HIV-1 replication in both human CD4+ T cells and HIV-infected humanized mice [128,240]. HIV infection also leads to lipid accumulation and increased OXPHOS in HIV-infected macrophages that use humanized mouse model [241]. HIV-infected humanized mice also showed gut barrier dysfunction, and elevated plasma and gut tissue oxidized lipoproteins [234]. Our collaborative studies demonstrated that a treatment (apolipoprotein A-I mimetic synthetic peptides designed to mimic apolipoprotein; and A-1 to remove excess cholesterol) could attenuate macrophage activation, and reduce systemic and gut inflammation in chronically treated HIV in humanized mice [234,235]. With the recent development of the germ-free humanized mice model, additional studies are now seeking to investigate the contribution of resident microbiota to human specific pathogen infection, including HIV [242]. In summary, humanized mouse models have emerged as a versatile animal model that can be used to support mechanistic and preclinical studies of HIV infection and ART-related metabolic stress and T cell dysfunction, including studies of drug treatment, supplement treatment and genetic manipulation.

6. Using the Humanized Mice Model to Study the Function and Exhaustion of Engineered CAR T Cell Immunity against HIV

Humanized mouse models provide an ideal platform to evaluate the therapeutic efficacy of engineered immunity and have been widely used to test immunotherapies for HIV and cancer [243,244,245,246]. They include, but are not limited to, bNAbs treatment, checkpoint inhibitor blockade, cytokine treatment, NK cell, and T cell-based therapies that seek to improve anti-HIV immunity and clear infected cells. The humanized BLT mouse model is a particularly good model for the study of T cell-based therapies because it has a human thymus organoid, enabling natural T cell selection and development within the model. The development of chimeric antigen receptor (CAR T) cell therapies, which have emerged as a promising therapy in recent years because of tremendous success as a cancer treatment, was critically influenced by the humanized mice model. Anti-HIV CAR-T cells are genetically engineered T cells that specifically target antigens on the surface of HIV-infected cells [245]. Unlike cytotoxic T lymphocytes (CTLs), CAR T cells do not rely on the endogenous T cell receptor (TCR) for antigen recognition and can bypass some of the limitations of natural CTLs, such as major histocompatibility complex (MHC) restriction and downregulation by HIV; they also directly target conserved regions of the virus, making it harder for the virus to escape [245], and can be engineered to resist HIV infection [245]. The CARs best-suited to HIV are CD4-based CARs, whose antigen recognition domain is the extracellular domain of CD4, which enables the recognition of HIV gp120 on infected cells [247,248,249,250,251,252]. Others have also reported the effective anti-HIV activity of T cells engineered with CAR designs based on broad neutralizing antibodies [203,253,254,255].
We have used the BLT mouse model of HIV infection to evaluate the efficacy of HSPC-derived CAR-T therapy and closely examine engineered antigen-specific T cell responses. We demonstrated that HSPC-based CD4CAR therapy allowed long-term engraftment and development of functional anti-HIV CAR-T cells, which suppressed viral replication [199,256]. We also found that the HSC-derived CAR-T cells persisted for an extended period in both humanized mice and non-human primates (NHPs) (>2 years) [257], indicating the potential for long-term viral control. Studies of humanized mice have allowed the extensive selection and optimization of CAR designs, which has in turn demonstrated the potential for anti-HIV CAR-T cells to contribute to a HIV cure. These critical findings have in turn paved the way for multiple ongoing clinical trials of anti-HIV CAR therapy (ClinicalTrials.gov Identifier: NCT04648046, NCT05077527, NCT03240328).
Interestingly, in both the humanized mouse and NHP models, CAR T cells also develop exhaustion and lose their ability to control the viral replication [256,258]. Humanized mouse models are ideal model to test various strategies to boost the functions of CAR T cells and prevent immune exhaustion. For example, PD-1 checkpoint blockade may enhance the CTL activity of HIV-CAR T cells [259]. Research of cancer immunology has also provided many potential strategies that could be used to improve CAR-T cell function and T cell mediated control [260]. The most widely studied approach is the blocking of inhibitory receptors or the genetic reduction of the expression of inhibitory receptors, with the aim of enhancing CAR-T cell function [261].
Metabolic remodeling is emerging as a promising method to improve the metabolic fitness of T cells and prevent/restore CAR-T cells from exhaustion. The use of 4-1BB costimulatory receptors has been shown to promote mitochondria biogenesis and OXPHOS of T cells [262], and studies, both by us and other researchers, have shown that anti-HIV CAR T cells with 4-1BB costimulatory domain have superior persistence and anti-viral functions [200,256]. New studies of cancer immunotherapy also indicate that manipulating glucose metabolism may result in beneficial metabolic adaptations. For example, glucose-starved T cells upregulate AMPK activity, which enhances mitochondria respiration and fatty acid usage, resulting in these T cells demonstrating better functions and delaying tumor growth [165,263,264,265].
Additionally, the optimization of amino acid nutritional support, enhancement of mitochondrial function, and modulation of both immune and metabolic checkpoints have emerged as novel ways to boost CAR T therapy [263,266,267,268]. For instance, a recent study has shown that the mitochondrial enzyme isocitrate dehydrogenase 2 (IDH2) reduces carboxylate glutamine in CD8 T cells. Inhibiting IDH2 in CAR T cells does not impair proliferation nor affect the effector function of the T cells, but does promote memory T cell formation and enhance antitumor responses [269]. This is especially relevant to HIV CAR-T cell research because of the chronic nature of HIV infection, and the associated importance of long-term immune cell function and persistence in maintaining immune surveillance. Engineering approaches to overcome the exhaustion of CAR T cell therapy will therefore most likely involve a combination of strategies that target immune and metabolic pathways.

7. Conclusions

Much of the complex interplay between HIV infection, inflammation, immune cell metabolism, and immune exhaustion remains a mystery to researchers and, it is in this context that humanized mouse models have a particular value, as a powerful and versatile tool that can be used to model HIV pathogenesis and test potential therapeutics. Further research is needed to explore the impact of metabolic remodeling in helping to alleviate chronic inflammation, prevent exhaustion, and improve endogenous and engineered T cells responses. It is however critical to understand the limitation of humanized mouse models, as the choice of mouse strain and method of construction may impact the level of human immune reconstitution, development of cellular and humoral responses, basal metabolic rate, and GVHD. Additional studies are needed to further improve the model so that it better recapitulates human conditions; this will in turn enable the investigation of the multiple factors that impact HIV immune pathogenesis, such as genetics, co-infections, gut microbiota, and immune metabolism.

Author Contributions

W.M. and A.Z. conceptualized this work. W.M., V.P. and A.Z. wrote this review together, S.K. reviewed and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the National Institute of Allergy and Infectious Diseases (grants 1R21AI140866 to AZ, R01AI172727 to AZ), the National Institute on Drug Abuse (grant R01DA-52841 to AZ), the Foundation for AIDS Research (amfAR) (grant 110304-71-RGRL, 110395-72-RPRL to AZ, 110038-67-RSRL to SGK), the National Cancer Institute (grant 1R01CA239261-01 to SGK); NIH grants P30AI28697 (to the UCLA CFAR Virology Core, Gene and Cell Therapy Core, and Humanized Mouse Core) and R01AI140775, U19AI149504 (to SGK); the California Institute for Regenerative Medicine (grant DISC2-10748) (to SGK); and California HIV/AIDS Research Program (grant H24BD7817, to WM). This work was also supported by the UCLA AIDS Institute, the James B. Pendleton Charitable Trust, and the McCarthy Family Foundation.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No specific data were generated to support reported results.

Conflicts of Interest

Donors had no role in the design of the study; the collection, analysis or interpretation of data; the writing of the manuscript; or the decision to publish the results.

References

  1. Deeks, S.G. HIV infection, inflammation, immunosenescence, and aging. Annu. Rev. Med. 2011, 62, 141–155. [Google Scholar] [CrossRef]
  2. Lv, T.; Cao, W.; Li, T. HIV-Related Immune Activation and Inflammation: Current Understanding and Strategies. J. Immunol. Res. 2021, 2021, 7316456. [Google Scholar] [CrossRef]
  3. Klatt, N.R.; Chomont, N.; Douek, D.C.; Deeks, S.G. Immune activation and HIV persistence: Implications for curative approaches to HIV infection. Immunol. Rev. 2013, 254, 326–342. [Google Scholar] [CrossRef] [PubMed]
  4. Trickey, A.; Sabin, C.A.; Burkholder, G.; Crane, H.; d’Arminio Monforte, A.; Egger, M.; Gill, M.J.; Grabar, S.; Guest, J.L.; Jarrin, I.; et al. Life expectancy after 2015 of adults with HIV on long-term antiretroviral therapy in Europe and North America: A collaborative analysis of cohort studies. Lancet HIV 2023, 10, e295–e307. [Google Scholar] [CrossRef]
  5. Antiretroviral Therapy Cohort, C. Survival of HIV-positive patients starting antiretroviral therapy between 1996 and 2013: A collaborative analysis of cohort studies. Lancet HIV 2017, 4, e349–e356. [Google Scholar] [CrossRef]
  6. Babu, H.; Ambikan, A.T.; Gabriel, E.E.; Svensson Akusjarvi, S.; Palaniappan, A.N.; Sundaraj, V.; Mupanni, N.R.; Sperk, M.; Cheedarla, N.; Sridhar, R.; et al. Systemic Inflammation and the Increased Risk of Inflamm-Aging and Age-Associated Diseases in People Living With HIV on Long Term Suppressive Antiretroviral Therapy. Front. Immunol. 2019, 10, 1965. [Google Scholar] [CrossRef]
  7. Hileman, C.O.; Funderburg, N.T. Inflammation, Immune Activation, and Antiretroviral Therapy in HIV. Curr. HIV/AIDS Rep. 2017, 14, 93–100. [Google Scholar] [CrossRef]
  8. Bloch, M.; John, M.; Smith, D.; Rasmussen, T.A.; Wright, E. Managing HIV-associated inflammation and ageing in the era of modern ART. HIV Med. 2020, 21 (Suppl. S3), 2–16. [Google Scholar] [CrossRef] [PubMed]
  9. Baker, J.V.; Hullsiek, K.H.; Singh, A.; Wilson, E.; Henry, K.; Lichtenstein, K.; Onen, N.; Kojic, E.; Patel, P.; Brooks, J.T.; et al. Immunologic predictors of coronary artery calcium progression in a contemporary HIV cohort. AIDS 2014, 28, 831–840. [Google Scholar] [CrossRef]
  10. Borges, A.H.; Silverberg, M.J.; Wentworth, D.; Grulich, A.E.; Fatkenheuer, G.; Mitsuyasu, R.; Tambussi, G.; Sabin, C.A.; Neaton, J.D.; Lundgren, J.D.; et al. Predicting risk of cancer during HIV infection: The role of inflammatory and coagulation biomarkers. AIDS 2013, 27, 1433–1441. [Google Scholar] [CrossRef]
  11. Burdo, T.H.; Lo, J.; Abbara, S.; Wei, J.; DeLelys, M.E.; Preffer, F.; Rosenberg, E.S.; Williams, K.C.; Grinspoon, S. Soluble CD163, a novel marker of activated macrophages, is elevated and associated with noncalcified coronary plaque in HIV-infected patients. J. Infect. Dis. 2011, 204, 1227–1236. [Google Scholar] [CrossRef]
  12. Subramanian, S.; Tawakol, A.; Burdo, T.H.; Abbara, S.; Wei, J.; Vijayakumar, J.; Corsini, E.; Abdelbaky, A.; Zanni, M.V.; Hoffmann, U.; et al. Arterial inflammation in patients with HIV. JAMA 2012, 308, 379–386. [Google Scholar] [CrossRef]
  13. Tenorio, A.R.; Zheng, Y.; Bosch, R.J.; Krishnan, S.; Rodriguez, B.; Hunt, P.W.; Plants, J.; Seth, A.; Wilson, C.C.; Deeks, S.G.; et al. Soluble markers of inflammation and coagulation but not T-cell activation predict non-AIDS-defining morbid events during suppressive antiretroviral treatment. J. Infect. Dis. 2014, 210, 1248–1259. [Google Scholar] [CrossRef]
  14. Nou, E.; Lo, J.; Grinspoon, S.K. Inflammation, immune activation, and cardiovascular disease in HIV. AIDS 2016, 30, 1495–1509. [Google Scholar] [CrossRef]
  15. Fenwick, C.; Joo, V.; Jacquier, P.; Noto, A.; Banga, R.; Perreau, M.; Pantaleo, G. T-cell exhaustion in HIV infection. Immunol. Rev. 2019, 292, 149–163. [Google Scholar] [CrossRef]
  16. Yates, K.B.; Tonnerre, P.; Martin, G.E.; Gerdemann, U.; Al Abosy, R.; Comstock, D.E.; Weiss, S.A.; Wolski, D.; Tully, D.C.; Chung, R.T.; et al. Epigenetic scars of CD8+ T cell exhaustion persist after cure of chronic infection in humans. Nat. Immunol. 2021, 22, 1020–1029. [Google Scholar] [CrossRef]
  17. Khaitan, A.; Unutmaz, D. Revisiting immune exhaustion during HIV infection. Curr. HIV/AIDS Rep. 2011, 8, 4–11. [Google Scholar] [CrossRef] [PubMed]
  18. Yu, Y.R.; Imrichova, H.; Wang, H.; Chao, T.; Xiao, Z.; Gao, M.; Rincon-Restrepo, M.; Franco, F.; Genolet, R.; Cheng, W.C.; et al. Disturbed mitochondrial dynamics in CD8+ TILs reinforce T cell exhaustion. Nat. Immunol. 2020, 21, 1540–1551. [Google Scholar] [CrossRef] [PubMed]
  19. Bengsch, B.; Johnson, A.L.; Kurachi, M.; Odorizzi, P.M.; Pauken, K.E.; Attanasio, J.; Stelekati, E.; McLane, L.M.; Paley, M.A.; Delgoffe, G.M.; et al. Bioenergetic Insufficiencies Due to Metabolic Alterations Regulated by the Inhibitory Receptor PD-1 Are an Early Driver of CD8+ T Cell Exhaustion. Immunity 2016, 45, 358–373. [Google Scholar] [CrossRef] [PubMed]
  20. Scharping, N.E.; Rivadeneira, D.B.; Menk, A.V.; Vignali, P.D.A.; Ford, B.R.; Rittenhouse, N.L.; Peralta, R.; Wang, Y.; Wang, Y.; DePeaux, K.; et al. Mitochondrial stress induced by continuous stimulation under hypoxia rapidly drives T cell exhaustion. Nat. Immunol. 2021, 22, 205–215. [Google Scholar] [CrossRef]
  21. Zheng, K.; Zheng, X.; Yang, W. The Role of Metabolic Dysfunction in T-Cell Exhaustion During Chronic Viral Infection. Front. Immunol. 2022, 13, 843242. [Google Scholar] [CrossRef]
  22. Zicari, S.; Sessa, L.; Cotugno, N.; Ruggiero, A.; Morrocchi, E.; Concato, C.; Rocca, S.; Zangari, P.; Manno, E.C.; Palma, P. Immune Activation, Inflammation, and Non-AIDS Co-Morbidities in HIV-Infected Patients under Long-Term ART. Viruses 2019, 11, 200. [Google Scholar] [CrossRef]
  23. Hsu, D.C.; Sereti, I. Serious Non-AIDS Events: Therapeutic Targets of Immune Activation and Chronic Inflammation in HIV Infection. Drugs 2016, 76, 533–549. [Google Scholar] [CrossRef]
  24. Hunt, P.W.; Martin, J.N.; Sinclair, E.; Bredt, B.; Hagos, E.; Lampiris, H.; Deeks, S.G. T cell activation is associated with lower CD4+ T cell gains in human immunodeficiency virus-infected patients with sustained viral suppression during antiretroviral therapy. J. Infect. Dis. 2003, 187, 1534–1543. [Google Scholar] [CrossRef] [PubMed]
  25. Kaplan, R.C.; Sinclair, E.; Landay, A.L.; Lurain, N.; Sharrett, A.R.; Gange, S.J.; Xue, X.; Hunt, P.; Karim, R.; Kern, D.M.; et al. T cell activation and senescence predict subclinical carotid artery disease in HIV-infected women. J. Infect. Dis. 2011, 203, 452–463. [Google Scholar] [CrossRef]
  26. Catalfamo, M.; Di Mascio, M.; Hu, Z.; Srinivasula, S.; Thaker, V.; Adelsberger, J.; Rupert, A.; Baseler, M.; Tagaya, Y.; Roby, G.; et al. HIV infection-associated immune activation occurs by two distinct pathways that differentially affect CD4 and CD8 T cells. Proc. Natl. Acad. Sci. USA 2008, 105, 19851–19856. [Google Scholar] [CrossRef]
  27. Hunt, P.W.; Brenchley, J.; Sinclair, E.; McCune, J.M.; Roland, M.; Page-Shafer, K.; Hsue, P.; Emu, B.; Krone, M.; Lampiris, H.; et al. Relationship between T cell activation and CD4+ T cell count in HIV-seropositive individuals with undetectable plasma HIV RNA levels in the absence of therapy. J. Infect. Dis. 2008, 197, 126–133. [Google Scholar] [CrossRef]
  28. Moir, S.; Fauci, A.S. B-cell exhaustion in HIV infection: The role of immune activation. Curr. Opin. HIV AIDS 2014, 9, 472–477. [Google Scholar] [CrossRef] [PubMed]
  29. Moir, S.; Fauci, A.S. B cells in HIV infection and disease. Nat. Rev. Immunol. 2009, 9, 235–245. [Google Scholar] [CrossRef] [PubMed]
  30. Martinez-Maza, O.; Breen, E.C. B-cell activation and lymphoma in patients with HIV. Curr. Opin. Oncol. 2002, 14, 528–532. [Google Scholar] [CrossRef]
  31. Castley, A.; Berry, C.; French, M.; Fernandez, S.; Krueger, R.; Nolan, D. Elevated plasma soluble CD14 and skewed CD16+ monocyte distribution persist despite normalisation of soluble CD163 and CXCL10 by effective HIV therapy: A changing paradigm for routine HIV laboratory monitoring? PLoS ONE 2014, 9, e115226. [Google Scholar] [CrossRef]
  32. Hearps, A.C.; Martin, G.E.; Angelovich, T.A.; Cheng, W.J.; Maisa, A.; Landay, A.L.; Jaworowski, A.; Crowe, S.M. Aging is associated with chronic innate immune activation and dysregulation of monocyte phenotype and function. Aging Cell 2012, 11, 867–875. [Google Scholar] [CrossRef]
  33. Hearps, A.C.; Maisa, A.; Cheng, W.J.; Angelovich, T.A.; Lichtfuss, G.F.; Palmer, C.S.; Landay, A.L.; Jaworowski, A.; Crowe, S.M. HIV infection induces age-related changes to monocytes and innate immune activation in young men that persist despite combination antiretroviral therapy. AIDS 2012, 26, 843–853. [Google Scholar] [CrossRef]
  34. Lichtfuss, G.F.; Hoy, J.; Rajasuriar, R.; Kramski, M.; Crowe, S.M.; Lewin, S.R. Biomarkers of immune dysfunction following combination antiretroviral therapy for HIV infection. Biomark. Med. 2011, 5, 171–186. [Google Scholar] [CrossRef]
  35. Rodger, A.J.; Fox, Z.; Lundgren, J.D.; Kuller, L.H.; Boesecke, C.; Gey, D.; Skoutelis, A.; Goetz, M.B.; Phillips, A.N.; INSIGHT Strategies for Management of Antiretroviral Therapy (SMART) Study Group. Activation and coagulation biomarkers are independent predictors of the development of opportunistic disease in patients with HIV infection. J. Infect. Dis. 2009, 200, 973–983. [Google Scholar] [CrossRef]
  36. Kuller, L.H.; Tracy, R.; Belloso, W.; De Wit, S.; Drummond, F.; Lane, H.C.; Ledergerber, B.; Lundgren, J.; Neuhaus, J.; Nixon, D.; et al. Inflammatory and coagulation biomarkers and mortality in patients with HIV infection. PLoS Med. 2008, 5, e203. [Google Scholar] [CrossRef]
  37. Ledwaba, L.; Tavel, J.A.; Khabo, P.; Maja, P.; Qin, J.; Sangweni, P.; Liu, X.; Follmann, D.; Metcalf, J.A.; Orsega, S.; et al. Pre-ART levels of inflammation and coagulation markers are strong predictors of death in a South African cohort with advanced HIV disease. PLoS ONE 2012, 7, e24243. [Google Scholar] [CrossRef]
  38. Wonderlich, E.R.; Subramanian, K.; Cox, B.; Wiegand, A.; Lackman-Smith, C.; Bale, M.J.; Stone, M.; Hoh, R.; Kearney, M.F.; Maldarelli, F.; et al. Effector memory differentiation increases detection of replication-competent HIV-l in resting CD4+ T cells from virally suppressed individuals. PLoS Pathog. 2019, 15, e1008074. [Google Scholar] [CrossRef]
  39. Vandergeeten, C.; Fromentin, R.; DaFonseca, S.; Lawani, M.B.; Sereti, I.; Lederman, M.M.; Ramgopal, M.; Routy, J.P.; Sekaly, R.P.; Chomont, N. Interleukin-7 promotes HIV persistence during antiretroviral therapy. Blood 2013, 121, 4321–4329. [Google Scholar] [CrossRef]
  40. Brooks, D.G.; Hamer, D.H.; Arlen, P.A.; Gao, L.; Bristol, G.; Kitchen, C.M.; Berger, E.A.; Zack, J.A. Molecular characterization, reactivation, and depletion of latent HIV. Immunity 2003, 19, 413–423. [Google Scholar] [CrossRef] [PubMed]
  41. Fletcher, C.V.; Staskus, K.; Wietgrefe, S.W.; Rothenberger, M.; Reilly, C.; Chipman, J.G.; Beilman, G.J.; Khoruts, A.; Thorkelson, A.; Schmidt, T.E.; et al. Persistent HIV-1 replication is associated with lower antiretroviral drug concentrations in lymphatic tissues. Proc. Natl. Acad. Sci. USA 2014, 111, 2307–2312. [Google Scholar] [CrossRef] [PubMed]
  42. Ganor, Y.; Real, F.; Sennepin, A.; Dutertre, C.A.; Prevedel, L.; Xu, L.; Tudor, D.; Charmeteau, B.; Couedel-Courteille, A.; Marion, S.; et al. HIV-1 reservoirs in urethral macrophages of patients under suppressive antiretroviral therapy. Nat. Microbiol. 2019, 4, 633–644. [Google Scholar] [CrossRef] [PubMed]
  43. Margolis, D.M.; Archin, N.M.; Cohen, M.S.; Eron, J.J.; Ferrari, G.; Garcia, J.V.; Gay, C.L.; Goonetilleke, N.; Joseph, S.B.; Swanstrom, R.; et al. Curing HIV: Seeking to Target and Clear Persistent Infection. Cell 2020, 181, 189–206. [Google Scholar] [CrossRef] [PubMed]
  44. Massanella, M.; Fromentin, R.; Chomont, N. Residual inflammation and viral reservoirs: Alliance against an HIV cure. Curr. Opin. HIV AIDS 2016, 11, 234–241. [Google Scholar] [CrossRef]
  45. Hunt, P.W.; Martin, J.N.; Sinclair, E.; Epling, L.; Teague, J.; Jacobson, M.A.; Tracy, R.P.; Corey, L.; Deeks, S.G. Valganciclovir reduces T cell activation in HIV-infected individuals with incomplete CD4+ T cell recovery on antiretroviral therapy. J. Infect. Dis. 2011, 203, 1474–1483. [Google Scholar] [CrossRef]
  46. Nazli, A.; Kafka, J.K.; Ferreira, V.H.; Anipindi, V.; Mueller, K.; Osborne, B.J.; Dizzell, S.; Chauvin, S.; Mian, M.F.; Ouellet, M.; et al. HIV-1 gp120 induces TLR2- and TLR4-mediated innate immune activation in human female genital epithelium. J. Immunol. 2013, 191, 4246–4258. [Google Scholar] [CrossRef]
  47. Ben Haij, N.; Planes, R.; Leghmari, K.; Serrero, M.; Delobel, P.; Izopet, J.; BenMohamed, L.; Bahraoui, E. HIV-1 Tat Protein Induces Production of Proinflammatory Cytokines by Human Dendritic Cells and Monocytes/Macrophages through Engagement of TLR4-MD2-CD14 Complex and Activation of NF-kappaB Pathway. PLoS ONE 2015, 10, e0129425. [Google Scholar] [CrossRef]
  48. Sarkar, R.; Mitra, D.; Chakrabarti, S. HIV-1 gp120 protein downregulates Nef induced IL-6 release in immature dentritic cells through interplay of DC-SIGN. PLoS ONE 2013, 8, e59073. [Google Scholar] [CrossRef]
  49. Yang, Y.; Wu, J.; Lu, Y. Mechanism of HIV-1-TAT induction of interleukin-1beta from human monocytes: Involvement of the phospholipase C/protein kinase C signaling cascade. J. Med. Virol. 2010, 82, 735–746. [Google Scholar] [CrossRef]
  50. Brigino, E.; Haraguchi, S.; Koutsonikolis, A.; Cianciolo, G.J.; Owens, U.; Good, R.A.; Day, N.K. Interleukin 10 is induced by recombinant HIV-1 Nef protein involving the calcium/calmodulin-dependent phosphodiesterase signal transduction pathway. Proc. Natl. Acad. Sci. USA 1997, 94, 3178–3182. [Google Scholar] [CrossRef] [PubMed]
  51. Ivashkiv, L.B.; Donlin, L.T. Regulation of type I interferon responses. Nat. Rev. Immunol. 2014, 14, 36–49. [Google Scholar] [CrossRef]
  52. Soper, A.; Kimura, I.; Nagaoka, S.; Konno, Y.; Yamamoto, K.; Koyanagi, Y.; Sato, K. Type I Interferon Responses by HIV-1 Infection: Association with Disease Progression and Control. Front. Immunol. 2017, 8, 1823. [Google Scholar] [CrossRef]
  53. Utay, N.S.; Douek, D.C. Interferons and HIV Infection: The Good, the Bad, and the Ugly. Pathog. Immun. 2016, 1, 107–116. [Google Scholar] [CrossRef]
  54. Doyle, T.; Goujon, C.; Malim, M.H. HIV-1 and interferons: Who’s interfering with whom? Nat. Rev. Microbiol. 2015, 13, 403–413. [Google Scholar] [CrossRef] [PubMed]
  55. Cabral-Piccin, M.P.; Papagno, L.; Lahaye, X.; Perdomo-Celis, F.; Volant, S.; White, E.; Monceaux, V.; Llewellyn-Lacey, S.; Fromentin, R.; Price, D.A.; et al. Primary role of type I interferons for the induction of functionally optimal antigen-specific CD8+ T cells in HIV infection. eBioMedicine 2023, 91, 104557. [Google Scholar] [CrossRef]
  56. Gondim, M.V.P.; Sherrill-Mix, S.; Bibollet-Ruche, F.; Russell, R.M.; Trimboli, S.; Smith, A.G.; Li, Y.; Liu, W.; Avitto, A.N.; DeVoto, J.C.; et al. Heightened resistance to host type 1 interferons characterizes HIV-1 at transmission and after antiretroviral therapy interruption. Sci. Transl. Med. 2021, 13, eabd8179. [Google Scholar] [CrossRef]
  57. Sandstrom, T.S.; Ranganath, N.; Angel, J.B. Impairment of the type I interferon response by HIV-1: Potential targets for HIV eradication. Cytokine Growth Factor Rev. 2017, 37, 1–16. [Google Scholar] [CrossRef] [PubMed]
  58. Barrat, F.J.; Crow, M.K.; Ivashkiv, L.B. Interferon target-gene expression and epigenomic signatures in health and disease. Nat. Immunol. 2019, 20, 1574–1583. [Google Scholar] [CrossRef]
  59. Snell, L.M.; McGaha, T.L.; Brooks, D.G. Type I Interferon in Chronic Virus Infection and Cancer. Trends Immunol. 2017, 38, 542–557. [Google Scholar] [CrossRef] [PubMed]
  60. Mackelprang, R.D.; Filali-Mouhim, A.; Richardson, B.; Lefebvre, F.; Katabira, E.; Ronald, A.; Gray, G.; Cohen, K.W.; Klatt, N.R.; Pecor, T.; et al. Upregulation of IFN-stimulated genes persists beyond the transitory broad immunologic changes of acute HIV-1 infection. iScience 2023, 26, 106454. [Google Scholar] [CrossRef]
  61. Su, L. Pathogenic Role of Type I Interferons in HIV-Induced Immune Impairments in Humanized Mice. Curr. HIV/AIDS Rep. 2019, 16, 224–229. [Google Scholar] [CrossRef]
  62. Swainson, L.A.; Sharma, A.A.; Ghneim, K.; Ribeiro, S.P.; Wilkinson, P.; Dunham, R.M.; Albright, R.G.; Wong, S.; Estes, J.D.; Piatak, M.; et al. IFN-α blockade during ART-treated SIV infection lowers tissue vDNA, rescues immune function, and improves overall health. JCI Insight 2022, 7, e153046. [Google Scholar] [CrossRef]
  63. Thaney, V.E.; Kaul, M. Type I Interferons in NeuroHIV. Viral Immunol. 2019, 32, 7–14. [Google Scholar] [CrossRef] [PubMed]
  64. Cheng, L.; Ma, J.; Li, J.; Li, D.; Li, G.; Li, F.; Zhang, Q.; Yu, H.; Yasui, F.; Ye, C.; et al. Blocking type I interferon signaling enhances T cell recovery and reduces HIV-1 reservoirs. J. Clin. Investig. 2017, 127, 269–279. [Google Scholar] [CrossRef]
  65. Zhen, A.; Rezek, V.; Youn, C.; Lam, B.; Chang, N.; Rick, J.; Carrillo, M.; Martin, H.; Kasparian, S.; Syed, P.; et al. Targeting type I interferon-mediated activation restores immune function in chronic HIV infection. J. Clin. Investig. 2017, 127, 260–268. [Google Scholar] [CrossRef]
  66. Deeks, S.G.; Odorizzi, P.M.; Sekaly, R.P. The interferon paradox: Can inhibiting an antiviral mechanism advance an HIV cure? J. Clin. Investig. 2017, 127, 103–105. [Google Scholar] [CrossRef]
  67. Klatt, N.R.; Funderburg, N.T.; Brenchley, J.M. Microbial translocation, immune activation, and HIV disease. Trends Microbiol. 2013, 21, 6–13. [Google Scholar] [CrossRef] [PubMed]
  68. Sandler, N.G.; Douek, D.C. Microbial translocation in HIV infection: Causes, consequences and treatment opportunities. Nat. Rev. Microbiol. 2012, 10, 655–666. [Google Scholar] [CrossRef] [PubMed]
  69. Marchetti, G.; Tincati, C.; Silvestri, G. Microbial translocation in the pathogenesis of HIV infection and AIDS. Clin. Microbiol. Rev. 2013, 26, 2–18. [Google Scholar] [CrossRef]
  70. Sharan, R.; Bucsan, A.N.; Ganatra, S.; Paiardini, M.; Mohan, M.; Mehra, S.; Khader, S.A.; Kaushal, D. Chronic Immune Activation in TB/HIV Co-infection. Trends Microbiol. 2020, 28, 619–632. [Google Scholar] [CrossRef]
  71. Patel, E.U.; Gianella, S.; Newell, K.; Tobian, A.A.; Kirkpatrick, A.R.; Nalugoda, F.; Grabowski, M.K.; Gray, R.H.; Serwadda, D.; Quinn, T.C.; et al. Elevated cytomegalovirus IgG antibody levels are associated with HIV-1 disease progression and immune activation. AIDS 2017, 31, 807–813. [Google Scholar] [CrossRef] [PubMed]
  72. Boulougoura, A.; Sereti, I. HIV infection and immune activation: The role of coinfections. Curr. Opin. HIV AIDS 2016, 11, 191–200. [Google Scholar] [CrossRef] [PubMed]
  73. Sabin, C.A.; Lundgren, J.D. The natural history of HIV infection. Curr. Opin. HIV AIDS 2013, 8, 311–317. [Google Scholar] [CrossRef] [PubMed]
  74. Raposo, M.A.; Armiliato, G.N.A.; Guimaraes, N.S.; Caram, C.A.; Silveira, R.D.S.; Tupinambas, U. Metabolic disorders and cardiovascular risk in people living with HIV/AIDS without the use of antiretroviral therapy. Rev. Soc. Bras. Med. Trop. 2017, 50, 598–606. [Google Scholar] [CrossRef]
  75. Riddler, S.A.; Smit, E.; Cole, S.R.; Li, R.; Chmiel, J.S.; Dobs, A.; Palella, F.; Visscher, B.; Evans, R.; Kingsley, L.A. Impact of HIV infection and HAART on serum lipids in men. JAMA 2003, 289, 2978–2982. [Google Scholar] [CrossRef] [PubMed]
  76. Grunfeld, C.; Pang, M.; Doerrler, W.; Shigenaga, J.K.; Jensen, P.; Feingold, K.R. Lipids, lipoproteins, triglyceride clearance, and cytokines in human immunodeficiency virus infection and the acquired immunodeficiency syndrome. J. Clin. Endocrinol. Metab. 1992, 74, 1045–1052. [Google Scholar] [CrossRef]
  77. Fitzgerald, M.L.; Mujawar, Z.; Tamehiro, N. ABC transporters, atherosclerosis and inflammation. Atherosclerosis 2010, 211, 361–370. [Google Scholar] [CrossRef] [PubMed]
  78. Rasheed, S.; Yan, J.S.; Lau, A.; Chan, A.S. HIV replication enhances production of free fatty acids, low density lipoproteins and many key proteins involved in lipid metabolism: A proteomics study. PLoS ONE 2008, 3, e3003. [Google Scholar] [CrossRef]
  79. Funderburg, N.T.; Mehta, N.N. Lipid Abnormalities and Inflammation in HIV Inflection. Curr. HIV/AIDS Rep. 2016, 13, 218–225. [Google Scholar] [CrossRef]
  80. Padmapriyadarsini, C.; Shet, A.; Srinivasan, R.; Ramachandran, G.; Sanjeeva, G.N.; Devi, P.; Ramesh, K.; Bhavani, P.K.; Reddy, D.; Suresh, E.; et al. High Prevalence of Lipid Abnormalities and Insulin Resistance Among Antiretroviral Naive HIV-infected Children in India. Pediatr. Infect. Dis. J. 2018, 37, 253–257. [Google Scholar] [CrossRef]
  81. Brener, M.I.; Post, W.S.; Haberlen, S.A.; Zhang, L.; Palella, F.J., Jr.; Jacobson, L.P.; Dobs, A.S.; George, R.T.; Witt, M.D.; Budoff, M.; et al. Comparison of Insulin Resistance to Coronary Atherosclerosis in Human Immunodeficiency Virus Infected and Uninfected Men (from the Multicenter AIDS Cohort Study). Am. J. Cardiol. 2016, 117, 993–1000. [Google Scholar] [CrossRef]
  82. Pedro, M.N.; Rocha, G.Z.; Guadagnini, D.; Santos, A.; Magro, D.O.; Assalin, H.B.; Oliveira, A.G.; Pedro, R.J.; Saad, M.J.A. Insulin Resistance in HIV-Patients: Causes and Consequences. Front. Endocrinol. 2018, 9, 514. [Google Scholar] [CrossRef] [PubMed]
  83. Mitchell, B.I.; Laws, E.I.; Chow, D.C.; SahBandar, I.N.; Gangcuangco, L.M.A.; Shikuma, C.M.; Ndhlovu, L.C. Increased Monocyte Inflammatory Responses to Oxidized LDL Are Associated with Insulin Resistance in HIV-Infected Individuals on Suppressive Antiretroviral Therapy. Viruses 2020, 12, 1129. [Google Scholar] [CrossRef]
  84. Sviridov, D.; Mukhamedova, N.; Makarov, A.A.; Adzhubei, A.; Bukrinsky, M. Comorbidities of HIV infection: Role of Nef-induced impairment of cholesterol metabolism and lipid raft functionality. AIDS 2020, 34, 1–13. [Google Scholar] [CrossRef]
  85. Brunham, L.R.; Kruit, J.K.; Pape, T.D.; Timmins, J.M.; Reuwer, A.Q.; Vasanji, Z.; Marsh, B.J.; Rodrigues, B.; Johnson, J.D.; Parks, J.S.; et al. Beta-cell ABCA1 influences insulin secretion, glucose homeostasis and response to thiazolidinedione treatment. Nat. Med. 2007, 13, 340–347. [Google Scholar] [CrossRef] [PubMed]
  86. Ergin, H.E.; Inga, E.E.; Maung, T.Z.; Javed, M.; Khan, S. HIV, Antiretroviral Therapy and Metabolic Alterations: A Review. Cureus 2020, 12, e8059. [Google Scholar] [CrossRef]
  87. Schank, M.; Zhao, J.; Moorman, J.P.; Yao, Z.Q. The Impact of HIV- and ART-Induced Mitochondrial Dysfunction in Cellular Senescence and Aging. Cells 2021, 10, 174. [Google Scholar] [CrossRef] [PubMed]
  88. Apostolova, N.; Blas-Garcia, A.; Esplugues, J.V. Mitochondrial interference by anti-HIV drugs: Mechanisms beyond Pol-γ inhibition. Trends Pharmacol. Sci. 2011, 32, 715–725. [Google Scholar] [CrossRef]
  89. Willig, A.L.; Overton, E.T. Metabolic Complications and Glucose Metabolism in HIV Infection: A Review of the Evidence. Curr. HIV/AIDS Rep. 2016, 13, 289–296. [Google Scholar] [CrossRef]
  90. Hruz, P.W. HIV protease inhibitors and insulin resistance: Lessons from in-vitro, rodent and healthy human volunteer models. Curr. Opin. HIV AIDS 2008, 3, 660–665. [Google Scholar] [CrossRef]
  91. Zhou, H.; Gurley, E.C.; Jarujaron, S.; Ding, H.; Fang, Y.; Xu, Z.; Pandak, W.M., Jr.; Hylemon, P.B. HIV protease inhibitors activate the unfolded protein response and disrupt lipid metabolism in primary hepatocytes. Am. J. Physiol. Gastrointest. Liver Physiol. 2006, 291, G1071–G1080. [Google Scholar] [CrossRef]
  92. Lewis, W.; Day, B.J.; Copeland, W.C. Mitochondrial toxicity of NRTI antiviral drugs: An integrated cellular perspective. Nat. Rev. Drug Discov. 2003, 2, 812–822. [Google Scholar] [CrossRef]
  93. Miserez, A.R.; Muller, P.Y.; Spaniol, V. Indinavir inhibits sterol-regulatory element-binding protein-1c-dependent lipoprotein lipase and fatty acid synthase gene activations. AIDS 2002, 16, 1587–1594. [Google Scholar] [CrossRef]
  94. Apostolova, N.; Gomez-Sucerquia, L.J.; Moran, A.; Alvarez, A.; Blas-Garcia, A.; Esplugues, J.V. Enhanced oxidative stress and increased mitochondrial mass during efavirenz-induced apoptosis in human hepatic cells. Br. J. Pharmacol. 2010, 160, 2069–2084. [Google Scholar] [CrossRef]
  95. Margolis, A.M.; Heverling, H.; Pham, P.A.; Stolbach, A. A review of the toxicity of HIV medications. J. Med. Toxicol. 2014, 10, 26–39. [Google Scholar] [CrossRef] [PubMed]
  96. Venhoff, N.; Setzer, B.; Melkaoui, K.; Walker, U.A. Mitochondrial toxicity of tenofovir, emtricitabine and abacavir alone and in combination with additional nucleoside reverse transcriptase inhibitors. Antivir. Ther. 2007, 12, 1075–1085. [Google Scholar] [CrossRef]
  97. McComsey, G.A.; Daar, E.S.; O’Riordan, M.; Collier, A.C.; Kosmiski, L.; Santana, J.L.; Fichtenbaum, C.J.; Fink, H.; Sax, P.E.; Libutti, D.E.; et al. Changes in fat mitochondrial DNA and function in subjects randomized to abacavir-lamivudine or tenofovir DF-emtricitabine with atazanavir-ritonavir or efavirenz: AIDS Clinical Trials Group study A5224s, substudy of A5202. J. Infect. Dis. 2013, 207, 604–611. [Google Scholar] [CrossRef]
  98. Hu, C.; Xuan, Y.; Zhang, X.; Liu, Y.; Yang, S.; Yang, K. Immune cell metabolism and metabolic reprogramming. Mol. Biol. Rep. 2022, 49, 9783–9795. [Google Scholar] [CrossRef]
  99. Domblides, C.; Lartigue, L.; Faustin, B. Metabolic Stress in the Immune Function of T Cells, Macrophages and Dendritic Cells. Cells 2018, 7, 68. [Google Scholar] [CrossRef]
  100. Li, S.Y.; Yin, L.B.; Ding, H.B.; Liu, M.; Lv, J.N.; Li, J.Q.; Wang, J.; Tang, T.; Fu, Y.J.; Jiang, Y.J.; et al. Altered lipid metabolites accelerate early dysfunction of T cells in HIV-infected rapid progressors by impairing mitochondrial function. Front. Immunol. 2023, 14, 1106881. [Google Scholar] [CrossRef]
  101. Ahmed, D.; Roy, D.; Cassol, E. Examining Relationships between Metabolism and Persistent Inflammation in HIV Patients on Antiretroviral Therapy. Mediat. Inflamm. 2018, 2018, 6238978. [Google Scholar] [CrossRef]
  102. Koethe, J.R.; Hulgan, T.; Niswender, K. Adipose tissue and immune function: A review of evidence relevant to HIV infection. J. Infect. Dis. 2013, 208, 1194–1201. [Google Scholar] [CrossRef]
  103. Bantug, G.R.; Galluzzi, L.; Kroemer, G.; Hess, C. The spectrum of T cell metabolism in health and disease. Nat. Rev. Immunol. 2018, 18, 19–34. [Google Scholar] [CrossRef]
  104. Dimeloe, S.; Burgener, A.V.; Grahlert, J.; Hess, C. T-cell metabolism governing activation, proliferation and differentiation; a modular view. Immunology 2017, 150, 35–44. [Google Scholar] [CrossRef]
  105. Zhang, W.; Xu, L.; Zhu, L.; Liu, Y.; Yang, S.; Zhao, M. Lipid Droplets, the Central Hub Integrating Cell Metabolism and the Immune System. Front. Physiol. 2021, 12, 746749. [Google Scholar] [CrossRef]
  106. Monson, E.A.; Trenerry, A.M.; Laws, J.L.; Mackenzie, J.M.; Helbig, K.J. Lipid droplets and lipid mediators in viral infection and immunity. FEMS Microbiol. Rev. 2021, 45, fuaa066. [Google Scholar] [CrossRef]
  107. Hara, Y.; Miura, S.; Komoto, S.; Inamura, T.; Koseki, S.; Watanabe, C.; Hokari, R.; Tsuzuki, Y.; Ogino, T.; Nagata, H.; et al. Exposure to fatty acids modulates interferon production by intraepithelial lymphocytes. Immunol. Lett. 2003, 86, 139–148. [Google Scholar] [CrossRef]
  108. Yan, J.; Horng, T. Lipid Metabolism in Regulation of Macrophage Functions. Trends Cell Biol. 2020, 30, 979–989. [Google Scholar] [CrossRef] [PubMed]
  109. York, A.G.; Williams, K.J.; Argus, J.P.; Zhou, Q.D.; Brar, G.; Vergnes, L.; Gray, E.E.; Zhen, A.; Wu, N.C.; Yamada, D.H.; et al. Limiting Cholesterol Biosynthetic Flux Spontaneously Engages Type I IFN Signaling. Cell 2015, 163, 1716–1729. [Google Scholar] [CrossRef]
  110. Crowe, S.M.; Westhorpe, C.L.; Mukhamedova, N.; Jaworowski, A.; Sviridov, D.; Bukrinsky, M. The macrophage: The intersection between HIV infection and atherosclerosis. J. Leukoc. Biol. 2010, 87, 589–598. [Google Scholar] [CrossRef]
  111. Willemsen, L.; Chen, H.J.; van Roomen, C.; Griffith, G.R.; Siebeler, R.; Neele, A.E.; Kroon, J.; Hoeksema, M.A.; de Winther, M.P.J. Monocyte and Macrophage Lipid Accumulation Results in Down-Regulated Type-I Interferon Responses. Front. Cardiovasc. Med. 2022, 9, 829877. [Google Scholar] [CrossRef]
  112. Hu, M.M.; Shu, H.B. Mitochondrial DNA-triggered innate immune response: Mechanisms and diseases. Cell. Mol. Immunol. 2023, 20, 1403–1412. [Google Scholar] [CrossRef] [PubMed]
  113. Marchi, S.; Guilbaud, E.; Tait, S.W.G.; Yamazaki, T.; Galluzzi, L. Mitochondrial control of inflammation. Nat. Rev. Immunol. 2023, 23, 159–173. [Google Scholar] [CrossRef]
  114. Jung, J.; Zeng, H.; Horng, T. Metabolism as a guiding force for immunity. Nat. Cell Biol. 2019, 21, 85–93. [Google Scholar] [CrossRef]
  115. Teer, E.; Mukonowenzou, N.C.; Essop, M.F. The Role of Immunometabolism in HIV-1 Pathogenicity: Links to Immune Cell Responses. Viruses 2022, 14, 1813. [Google Scholar] [CrossRef]
  116. Wherry, E.J.; Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 2015, 15, 486–499. [Google Scholar] [CrossRef]
  117. Kaufmann, D.E.; Kavanagh, D.G.; Pereyra, F.; Zaunders, J.J.; Mackey, E.W.; Miura, T.; Palmer, S.; Brockman, M.; Rathod, A.; Piechocka-Trocha, A.; et al. Upregulation of CTLA-4 by HIV-specific CD4+ T cells correlates with disease progression and defines a reversible immune dysfunction. Nat. Immunol. 2007, 8, 1246–1254. [Google Scholar] [CrossRef] [PubMed]
  118. Day, C.L.; Kaufmann, D.E.; Kiepiela, P.; Brown, J.A.; Moodley, E.S.; Reddy, S.; Mackey, E.W.; Miller, J.D.; Leslie, A.J.; DePierres, C.; et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 2006, 443, 350–354. [Google Scholar] [CrossRef]
  119. El-Far, M.; Halwani, R.; Said, E.; Trautmann, L.; Doroudchi, M.; Janbazian, L.; Fonseca, S.; van Grevenynghe, J.; Yassine-Diab, B.; Sekaly, R.P.; et al. T-cell exhaustion in HIV infection. Curr. HIV/AIDS Rep. 2008, 5, 13–19. [Google Scholar] [CrossRef] [PubMed]
  120. Attanasio, J.; Wherry, E.J. Costimulatory and Coinhibitory Receptor Pathways in Infectious Disease. Immunity 2016, 44, 1052–1068. [Google Scholar] [CrossRef]
  121. Wherry, E.J.; Ha, S.J.; Kaech, S.M.; Haining, W.N.; Sarkar, S.; Kalia, V.; Subramaniam, S.; Blattman, J.N.; Barber, D.L.; Ahmed, R. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 2007, 27, 670–684. [Google Scholar] [CrossRef] [PubMed]
  122. Saez-Cirion, A.; Sereti, I. Immunometabolism and HIV-1 pathogenesis: Food for thought. Nat. Rev. Immunol. 2021, 21, 5–19. [Google Scholar] [CrossRef] [PubMed]
  123. Leone, R.D.; Powell, J.D. Metabolism of immune cells in cancer. Nat. Rev. Cancer 2020, 20, 516–531. [Google Scholar] [CrossRef] [PubMed]
  124. Alwarawrah, Y.; Kiernan, K.; MacIver, N.J. Changes in Nutritional Status Impact Immune Cell Metabolism and Function. Front. Immunol. 2018, 9, 1055. [Google Scholar] [CrossRef]
  125. Pearce, E.L.; Pearce, E.J. Metabolic pathways in immune cell activation and quiescence. Immunity 2013, 38, 633–643. [Google Scholar] [CrossRef]
  126. O’Neill, L.A.; Kishton, R.J.; Rathmell, J. A guide to immunometabolism for immunologists. Nat. Rev. Immunol. 2016, 16, 553–565. [Google Scholar] [CrossRef]
  127. Reina-Campos, M.; Scharping, N.E.; Goldrath, A.W. CD8+ T cell metabolism in infection and cancer. Nat. Rev. Immunol. 2021, 21, 718–738. [Google Scholar] [CrossRef]
  128. Guo, H.; Wang, Q.; Ghneim, K.; Wang, L.; Rampanelli, E.; Holley-Guthrie, E.; Cheng, L.; Garrido, C.; Margolis, D.M.; Eller, L.A.; et al. Multi-omics analyses reveal that HIV-1 alters CD4+ T cell immunometabolism to fuel virus replication. Nat. Immunol. 2021, 22, 423–433. [Google Scholar] [CrossRef]
  129. Wik, J.A.; Skalhegg, B.S. T Cell Metabolism in Infection. Front. Immunol. 2022, 13, 840610. [Google Scholar] [CrossRef]
  130. Palmer, C.S.; Cherry, C.L.; Sada-Ovalle, I.; Singh, A.; Crowe, S.M. Glucose Metabolism in T Cells and Monocytes: New Perspectives in HIV Pathogenesis. eBioMedicine 2016, 6, 31–41. [Google Scholar] [CrossRef]
  131. Deguit, C.D.T.; Hough, M.; Hoh, R.; Krone, M.; Pilcher, C.D.; Martin, J.N.; Deeks, S.G.; McCune, J.M.; Hunt, P.W.; Rutishauser, R.L. Some Aspects of CD8+ T-Cell Exhaustion Are Associated With Altered T-Cell Mitochondrial Features and ROS Content in HIV Infection. J. Acquir. Immune Defic. Syndr. 2019, 82, 211–219. [Google Scholar] [CrossRef] [PubMed]
  132. Couret, J.; Chang, T.L. Reactive Oxygen Species in HIV Infection. EC Microbiol. 2016, 3, 597–604. [Google Scholar]
  133. Gangcuangco, L.M.A.; Mitchell, B.I.; Siriwardhana, C.; Kohorn, L.B.; Chew, G.M.; Bowler, S.; Kallianpur, K.J.; Chow, D.C.; Ndhlovu, L.C.; Gerschenson, M.; et al. Mitochondrial oxidative phosphorylation in peripheral blood mononuclear cells is decreased in chronic HIV and correlates with immune dysregulation. PLoS ONE 2020, 15, e0231761. [Google Scholar] [CrossRef] [PubMed]
  134. Valle-Casuso, J.C.; Angin, M.; Volant, S.; Passaes, C.; Monceaux, V.; Mikhailova, A.; Bourdic, K.; Avettand-Fenoel, V.; Boufassa, F.; Sitbon, M.; et al. Cellular Metabolism Is a Major Determinant of HIV-1 Reservoir Seeding in CD4+ T Cells and Offers an Opportunity to Tackle Infection. Cell Metab. 2019, 29, 611–626.E5. [Google Scholar] [CrossRef] [PubMed]
  135. Datta, P.K.; Deshmane, S.; Khalili, K.; Merali, S.; Gordon, J.C.; Fecchio, C.; Barrero, C.A. Glutamate metabolism in HIV-1 infected macrophages: Role of HIV-1 Vpr. Cell Cycle 2016, 15, 2288–2298. [Google Scholar] [CrossRef]
  136. Palmer, C.S.; Anzinger, J.J.; Zhou, J.; Gouillou, M.; Landay, A.; Jaworowski, A.; McCune, J.M.; Crowe, S.M. Glucose transporter 1-expressing proinflammatory monocytes are elevated in combination antiretroviral therapy-treated and untreated HIV+ subjects. J. Immunol. 2014, 193, 5595–5603. [Google Scholar] [CrossRef]
  137. Loisel-Meyer, S.; Swainson, L.; Craveiro, M.; Oburoglu, L.; Mongellaz, C.; Costa, C.; Martinez, M.; Cosset, F.L.; Battini, J.L.; Herzenberg, L.A.; et al. Glut1-mediated glucose transport regulates HIV infection. Proc. Natl. Acad. Sci. USA 2012, 109, 2549–2554. [Google Scholar] [CrossRef]
  138. Hegedus, A.; Kavanagh Williamson, M.; Khan, M.B.; Dias Zeidler, J.; Da Poian, A.T.; El-Bacha, T.; Struys, E.A.; Huthoff, H. Evidence for Altered Glutamine Metabolism in Human Immunodeficiency Virus Type 1 Infected Primary Human CD4+ T Cells. AIDS Res. Hum. Retroviruses 2017, 33, 1236–1247. [Google Scholar] [CrossRef]
  139. Hollenbaugh, J.A.; Munger, J.; Kim, B. Metabolite profiles of human immunodeficiency virus infected CD4+ T cells and macrophages using LC-MS/MS analysis. Virology 2011, 415, 153–159. [Google Scholar] [CrossRef]
  140. Kavanagh Williamson, M.; Coombes, N.; Juszczak, F.; Athanasopoulos, M.; Khan, M.B.; Eykyn, T.R.; Srenathan, U.; Taams, L.S.; Dias Zeidler, J.; Da Poian, A.T.; et al. Upregulation of Glucose Uptake and Hexokinase Activity of Primary Human CD4+ T Cells in Response to Infection with HIV-1. Viruses 2018, 10, 114. [Google Scholar] [CrossRef] [PubMed]
  141. Palmer, C.S.; Ostrowski, M.; Gouillou, M.; Tsai, L.; Yu, D.; Zhou, J.; Henstridge, D.C.; Maisa, A.; Hearps, A.C.; Lewin, S.R.; et al. Increased glucose metabolic activity is associated with CD4+ T-cell activation and depletion during chronic HIV infection. AIDS 2014, 28, 297–309. [Google Scholar] [CrossRef]
  142. Wang, Y.Y.; Zhen, C.; Hu, W.; Huang, H.H.; Li, Y.J.; Zhou, M.J.; Li, J.; Fu, Y.L.; Zhang, P.; Li, X.Y.; et al. Elevated glutamate impedes anti-HIV-1 CD8 + T cell responses in HIV-1-infected individuals on antiretroviral therapy. Commun. Biol. 2023, 6, 696. [Google Scholar] [CrossRef]
  143. Palmer, C.S.; Hussain, T.; Duette, G.; Weller, T.J.; Ostrowski, M.; Sada-Ovalle, I.; Crowe, S.M. Regulators of Glucose Metabolism in CD4+ and CD8+ T Cells. Int. Rev. Immunol. 2016, 35, 477–488. [Google Scholar] [CrossRef]
  144. McKnight, T.R.; Yoshihara, H.A.; Sitole, L.J.; Martin, J.N.; Steffens, F.; Meyer, D. A combined chemometric and quantitative NMR analysis of HIV/AIDS serum discloses metabolic alterations associated with disease status. Mol. Biosyst. 2014, 10, 2889–2897. [Google Scholar] [CrossRef]
  145. Yang, L.; Chu, Z.; Liu, M.; Zou, Q.; Li, J.; Liu, Q.; Wang, Y.; Wang, T.; Xiang, J.; Wang, B. Amino acid metabolism in immune cells: Essential regulators of the effector functions, and promising opportunities to enhance cancer immunotherapy. J. Hematol. Oncol. 2023, 16, 59. [Google Scholar] [CrossRef]
  146. Mondanelli, G.; Iacono, A.; Allegrucci, M.; Puccetti, P.; Grohmann, U. Immunoregulatory Interplay Between Arginine and Tryptophan Metabolism in Health and Disease. Front. Immunol. 2019, 10, 1565. [Google Scholar] [CrossRef] [PubMed]
  147. Mbongue, J.C.; Nicholas, D.A.; Torrez, T.W.; Kim, N.S.; Firek, A.F.; Langridge, W.H. The Role of Indoleamine 2, 3-Dioxygenase in Immune Suppression and Autoimmunity. Vaccines 2015, 3, 703–729. [Google Scholar] [CrossRef] [PubMed]
  148. Bipath, P.; Levay, P.F.; Viljoen, M. The kynurenine pathway activities in a sub-Saharan HIV/AIDS population. BMC Infect. Dis. 2015, 15, 346. [Google Scholar] [CrossRef] [PubMed]
  149. Favre, D.; Mold, J.; Hunt, P.W.; Kanwar, B.; Loke, P.; Seu, L.; Barbour, J.D.; Lowe, M.M.; Jayawardene, A.; Aweeka, F.; et al. Tryptophan catabolism by indoleamine 2,3-dioxygenase 1 alters the balance of TH17 to regulatory T cells in HIV disease. Sci. Transl. Med. 2010, 2, 32ra36. [Google Scholar] [CrossRef] [PubMed]
  150. Patsoukis, N.; Bardhan, K.; Chatterjee, P.; Sari, D.; Liu, B.; Bell, L.N.; Karoly, E.D.; Freeman, G.J.; Petkova, V.; Seth, P.; et al. PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation. Nat. Commun. 2015, 6, 6692. [Google Scholar] [CrossRef] [PubMed]
  151. Li, F.; Liu, H.; Zhang, D.; Ma, Y.; Zhu, B. Metabolic plasticity and regulation of T cell exhaustion. Immunology 2022, 167, 482–494. [Google Scholar] [CrossRef] [PubMed]
  152. Scharping, N.E.; Menk, A.V.; Moreci, R.S.; Whetstone, R.D.; Dadey, R.E.; Watkins, S.C.; Ferris, R.L.; Delgoffe, G.M. The Tumor Microenvironment Represses T Cell Mitochondrial Biogenesis to Drive Intratumoral T Cell Metabolic Insufficiency and Dysfunction. Immunity 2016, 45, 374–388. [Google Scholar] [CrossRef]
  153. Schurich, A.; Pallett, L.J.; Jajbhay, D.; Wijngaarden, J.; Otano, I.; Gill, U.S.; Hansi, N.; Kennedy, P.T.; Nastouli, E.; Gilson, R.; et al. Distinct Metabolic Requirements of Exhausted and Functional Virus-Specific CD8 T Cells in the Same Host. Cell Rep. 2016, 16, 1243–1252. [Google Scholar] [CrossRef] [PubMed]
  154. Staron, M.M.; Gray, S.M.; Marshall, H.D.; Parish, I.A.; Chen, J.H.; Perry, C.J.; Cui, G.; Li, M.O.; Kaech, S.M. The transcription factor FoxO1 sustains expression of the inhibitory receptor PD-1 and survival of antiviral CD8+ T cells during chronic infection. Immunity 2014, 41, 802–814. [Google Scholar] [CrossRef] [PubMed]
  155. Jin, H.S.; Suh, H.W.; Kim, S.J.; Jo, E.K. Mitochondrial Control of Innate Immunity and Inflammation. Immune Netw. 2017, 17, 77–88. [Google Scholar] [CrossRef]
  156. Bhaskaran, N.; Schneider, E.; Faddoul, F.; Paes da Silva, A.; Asaad, R.; Talla, A.; Greenspan, N.; Levine, A.D.; McDonald, D.; Karn, J.; et al. Oral immune dysfunction is associated with the expansion of FOXP3+PD-1+Amphiregulin+ T cells during HIV infection. Nat. Commun. 2021, 12, 5143. [Google Scholar] [CrossRef] [PubMed]
  157. Boasso, A.; Shearer, G.M.; Chougnet, C. Immune dysregulation in human immunodeficiency virus infection: Know it, fix it, prevent it? J. Intern. Med. 2009, 265, 78–96. [Google Scholar] [CrossRef]
  158. Faia, C.; Plaisance-Bonstaff, K.; Peruzzi, F. In vitro models of HIV-1 infection of the Central Nervous System. Drug Discov. Today Dis. Models 2020, 32, 5–11. [Google Scholar] [CrossRef]
  159. Sahu, G.K.; Lee, K.; Ji, J.; Braciale, V.; Baron, S.; Cloyd, M.W. A novel in vitro system to generate and study latently HIV-infected long-lived normal CD4+ T-lymphocytes. Virology 2006, 355, 127–137. [Google Scholar] [CrossRef]
  160. Kang, S.; Tang, H. HIV-1 Infection and Glucose Metabolism Reprogramming of T Cells: Another Approach Toward Functional Cure and Reservoir Eradication. Front. Immunol. 2020, 11, 572677. [Google Scholar] [CrossRef] [PubMed]
  161. Mercurio, V.; Fitzgerald, W.; Molodtsov, I.; Margolis, L. Persistent Immune Activation in HIV-1-Infected Ex Vivo Model Tissues Subjected to Antiretroviral Therapy: Soluble and Extracellular Vesicle-Associated Cytokines. J. Acquir. Immune Defic. Syndr. 2020, 84, 45–53. [Google Scholar] [CrossRef] [PubMed]
  162. Shi, D.; Mi, G.; Wang, M.; Webster, T.J. In vitro and ex vivo systems at the forefront of infection modeling and drug discovery. Biomaterials 2019, 198, 228–249. [Google Scholar] [CrossRef]
  163. Saez-Cirion, A.; Shin, S.Y.; Versmisse, P.; Barre-Sinoussi, F.; Pancino, G. Ex vivo T cell-based HIV suppression assay to evaluate HIV-specific CD8+ T-cell responses. Nat. Protoc. 2010, 5, 1033–1041. [Google Scholar] [CrossRef] [PubMed]
  164. Rahman, A.N.; Liu, J.; Mujib, S.; Kidane, S.; Ali, A.; Szep, S.; Han, C.; Bonner, P.; Parsons, M.; Benko, E.; et al. Elevated glycolysis imparts functional ability to CD8+ T cells in HIV infection. Life Sci. Alliance 2021, 4, e202101081. [Google Scholar] [CrossRef] [PubMed]
  165. Sukumar, M.; Liu, J.; Ji, Y.; Subramanian, M.; Crompton, J.G.; Yu, Z.; Roychoudhuri, R.; Palmer, D.C.; Muranski, P.; Karoly, E.D.; et al. Inhibiting glycolytic metabolism enhances CD8+ T cell memory and antitumor function. J. Clin. Investig. 2013, 123, 4479–4488. [Google Scholar] [CrossRef]
  166. Simon, V.; Ho, D.D.; Abdool Karim, Q. HIV/AIDS epidemiology, pathogenesis, prevention, and treatment. Lancet 2006, 368, 489–504. [Google Scholar] [CrossRef]
  167. Sereti, I.; Lane, H.C. Immunopathogenesis of human immunodeficiency virus: Implications for immune-based therapies. Clin. Infect. Dis. 2001, 32, 1738–1755. [Google Scholar] [CrossRef] [PubMed]
  168. Schmitz, J.E.; Korioth-Schmitz, B. Immunopathogenesis of simian immunodeficiency virus infection in nonhuman primates. Curr. Opin. HIV AIDS 2013, 8, 273–279. [Google Scholar] [CrossRef]
  169. Harris, L.D.; Tabb, B.; Sodora, D.L.; Paiardini, M.; Klatt, N.R.; Douek, D.C.; Silvestri, G.; Muller-Trutwin, M.; Vasile-Pandrea, I.; Apetrei, C.; et al. Downregulation of robust acute type I interferon responses distinguishes nonpathogenic simian immunodeficiency virus (SIV) infection of natural hosts from pathogenic SIV infection of rhesus macaques. J. Virol. 2010, 84, 7886–7891. [Google Scholar] [CrossRef]
  170. Jacquelin, B.; Mayau, V.; Targat, B.; Liovat, A.S.; Kunkel, D.; Petitjean, G.; Dillies, M.A.; Roques, P.; Butor, C.; Silvestri, G.; et al. Nonpathogenic SIV infection of African green monkeys induces a strong but rapidly controlled type I IFN response. J. Clin. Investig. 2009, 119, 3544–3555. [Google Scholar] [CrossRef] [PubMed]
  171. Estes, J.D.; Wong, S.W.; Brenchley, J.M. Nonhuman primate models of human viral infections. Nat. Rev. Immunol. 2018, 18, 390–404. [Google Scholar] [CrossRef]
  172. Garcia-Tellez, T.; Huot, N.; Ploquin, M.J.; Rascle, P.; Jacquelin, B.; Muller-Trutwin, M. Non-human primates in HIV research: Achievements, limits and alternatives. Infect. Genet. Evol. 2016, 46, 324–332. [Google Scholar] [CrossRef]
  173. Estes, J.D. Pathobiology of HIV/SIV-associated changes in secondary lymphoid tissues. Immunol. Rev. 2013, 254, 65–77. [Google Scholar] [CrossRef] [PubMed]
  174. Silvestri, G.; Paiardini, M.; Pandrea, I.; Lederman, M.M.; Sodora, D.L. Understanding the benign nature of SIV infection in natural hosts. J. Clin. Investig. 2007, 117, 3148–3154. [Google Scholar] [CrossRef] [PubMed]
  175. Brenchley, J.M.; Price, D.A.; Schacker, T.W.; Asher, T.E.; Silvestri, G.; Rao, S.; Kazzaz, Z.; Bornstein, E.; Lambotte, O.; Altmann, D.; et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat. Med. 2006, 12, 1365–1371. [Google Scholar] [CrossRef]
  176. Mylvaganam, G.H.; Chea, L.S.; Tharp, G.K.; Hicks, S.; Velu, V.; Iyer, S.S.; Deleage, C.; Estes, J.D.; Bosinger, S.E.; Freeman, G.J.; et al. Combination anti-PD-1 and antiretroviral therapy provides therapeutic benefit against SIV. JCI Insight 2018, 3, e153046. [Google Scholar] [CrossRef] [PubMed]
  177. Chew, G.M.; Fujita, T.; Webb, G.M.; Burwitz, B.J.; Wu, H.L.; Reed, J.S.; Hammond, K.B.; Clayton, K.L.; Ishii, N.; Abdel-Mohsen, M.; et al. TIGIT Marks Exhausted T Cells, Correlates with Disease Progression, and Serves as a Target for Immune Restoration in HIV and SIV Infection. PLoS Pathog. 2016, 12, e1005349. [Google Scholar] [CrossRef] [PubMed]
  178. Petrovas, C.; Price, D.A.; Mattapallil, J.; Ambrozak, D.R.; Geldmacher, C.; Cecchinato, V.; Vaccari, M.; Tryniszewska, E.; Gostick, E.; Roederer, M.; et al. SIV-specific CD8+ T cells express high levels of PD1 and cytokines but have impaired proliferative capacity in acute and chronic SIVmac251 infection. Blood 2007, 110, 928–936. [Google Scholar] [CrossRef] [PubMed]
  179. Velu, V.; Kannanganat, S.; Ibegbu, C.; Chennareddi, L.; Villinger, F.; Freeman, G.J.; Ahmed, R.; Amara, R.R. Elevated expression levels of inhibitory receptor programmed death 1 on simian immunodeficiency virus-specific CD8 T cells during chronic infection but not after vaccination. J. Virol. 2007, 81, 5819–5828. [Google Scholar] [CrossRef]
  180. Friedrich, T.C.; Valentine, L.E.; Yant, L.J.; Rakasz, E.G.; Piaskowski, S.M.; Furlott, J.R.; Weisgrau, K.L.; Burwitz, B.; May, G.E.; Leon, E.J.; et al. Subdominant CD8+ T-cell responses are involved in durable control of AIDS virus replication. J. Virol. 2007, 81, 3465–3476. [Google Scholar] [CrossRef] [PubMed]
  181. Jin, X.; Bauer, D.E.; Tuttleton, S.E.; Lewin, S.; Gettie, A.; Blanchard, J.; Irwin, C.E.; Safrit, J.T.; Mittler, J.; Weinberger, L.; et al. Dramatic rise in plasma viremia after CD8+ T cell depletion in simian immunodeficiency virus-infected macaques. J. Exp. Med. 1999, 189, 991–998. [Google Scholar] [CrossRef]
  182. Schmitz, J.E.; Kuroda, M.J.; Santra, S.; Sasseville, V.G.; Simon, M.A.; Lifton, M.A.; Racz, P.; Tenner-Racz, K.; Dalesandro, M.; Scallon, B.J.; et al. Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes. Science 1999, 283, 857–860. [Google Scholar] [CrossRef]
  183. Terrade, G.; Huot, N.; Petitdemange, C.; Lazzerini, M.; Orta Resendiz, A.; Jacquelin, B.; Muller-Trutwin, M. Interests of the Non-Human Primate Models for HIV Cure Research. Vaccines 2021, 9, 958. [Google Scholar] [CrossRef]
  184. Thippeshappa, R.; Kimata, J.T.; Kaushal, D. Toward a Macaque Model of HIV-1 Infection: Roadblocks, Progress, and Future Strategies. Front. Microbiol. 2020, 11, 882. [Google Scholar] [CrossRef] [PubMed]
  185. Bender, A.M.; Simonetti, F.R.; Kumar, M.R.; Fray, E.J.; Bruner, K.M.; Timmons, A.E.; Tai, K.Y.; Jenike, K.M.; Antar, A.A.R.; Liu, P.T.; et al. The Landscape of Persistent Viral Genomes in ART-Treated SIV, SHIV, and HIV-2 Infections. Cell Host Microbe 2019, 26, 73–85.e74. [Google Scholar] [CrossRef] [PubMed]
  186. Williams, K.C.; Burdo, T.H. HIV and SIV infection: The role of cellular restriction and immune responses in viral replication and pathogenesis. APMIS 2009, 117, 400–412. [Google Scholar] [CrossRef]
  187. Chuprin, J.; Buettner, H.; Seedhom, M.O.; Greiner, D.L.; Keck, J.G.; Ishikawa, F.; Shultz, L.D.; Brehm, M.A. Humanized mouse models for immuno-oncology research. Nat. Rev. Clin. Oncol. 2023, 20, 192–206. [Google Scholar] [CrossRef] [PubMed]
  188. Chen, J.; Liao, S.; Xiao, Z.; Pan, Q.; Wang, X.; Shen, K.; Wang, S.; Yang, L.; Guo, F.; Liu, H.F.; et al. The development and improvement of immunodeficient mice and humanized immune system mouse models. Front. Immunol. 2022, 13, 1007579. [Google Scholar] [CrossRef] [PubMed]
  189. Dash, P.K.; Gorantla, S.; Poluektova, L.; Hasan, M.; Waight, E.; Zhang, C.; Markovic, M.; Edagwa, B.; Machhi, J.; Olson, K.E.; et al. Humanized Mice for Infectious and Neurodegenerative disorders. Retrovirology 2021, 18, 13. [Google Scholar] [CrossRef] [PubMed]
  190. Allen, T.M.; Brehm, M.A.; Bridges, S.; Ferguson, S.; Kumar, P.; Mirochnitchenko, O.; Palucka, K.; Pelanda, R.; Sanders-Beer, B.; Shultz, L.D.; et al. Humanized immune system mouse models: Progress, challenges and opportunities. Nat. Immunol. 2019, 20, 770–774. [Google Scholar] [CrossRef] [PubMed]
  191. Victor Garcia, J. Humanized mice for HIV and AIDS research. Curr. Opin. Virol. 2016, 19, 56–64. [Google Scholar] [CrossRef]
  192. Terahara, K.; Iwabuchi, R.; Tsunetsugu-Yokota, Y. Perspectives on Non-BLT Humanized Mouse Models for Studying HIV Pathogenesis and Therapy. Viruses 2021, 13, 776. [Google Scholar] [CrossRef]
  193. Marsden, M.D. Benefits and limitations of humanized mice in HIV persistence studies. Retrovirology 2020, 17, 7. [Google Scholar] [CrossRef]
  194. Weichseldorfer, M.; Heredia, A.; Reitz, M.; Bryant, J.L.; Latinovic, O.S. Use of Humanized Mouse Models for Studying HIV-1 Infection, Pathogenesis and Persistence. J. AIDS HIV Treat. 2020, 2, 23–29. [Google Scholar]
  195. Abeynaike, S.; Paust, S. Humanized Mice for the Evaluation of Novel HIV-1 Therapies. Front. Immunol. 2021, 12, 636775. [Google Scholar] [CrossRef]
  196. Dudek, T.E.; Allen, T.M. HIV-specific CD8+ T-cell immunity in humanized bone marrow-liver-thymus mice. J. Infect. Dis. 2013, 208 (Suppl. 2), S150–S154. [Google Scholar] [CrossRef]
  197. Dudek, T.E.; No, D.C.; Seung, E.; Vrbanac, V.D.; Fadda, L.; Bhoumik, P.; Boutwell, C.L.; Power, K.A.; Gladden, A.D.; Battis, L.; et al. Rapid evolution of HIV-1 to functional CD8+ T cell responses in humanized BLT mice. Sci. Transl. Med. 2012, 4, 143ra198. [Google Scholar] [CrossRef] [PubMed]
  198. Karpel, M.E.; Boutwell, C.L.; Allen, T.M. BLT humanized mice as a small animal model of HIV infection. Curr. Opin. Virol. 2015, 13, 75–80. [Google Scholar] [CrossRef] [PubMed]
  199. Zhen, A.; Kamata, M.; Rezek, V.; Rick, J.; Levin, B.; Kasparian, S.; Chen, I.S.; Yang, O.O.; Zack, J.A.; Kitchen, S.G. HIV-specific Immunity Derived From Chimeric Antigen Receptor-engineered Stem Cells. Mol. Ther. 2015, 23, 1358–1367. [Google Scholar] [CrossRef]
  200. Maldini, C.R.; Claiborne, D.T.; Okawa, K.; Chen, T.; Dopkin, D.L.; Shan, X.; Power, K.A.; Trifonova, R.T.; Krupp, K.; Phelps, M.; et al. Dual CD4-based CAR T cells with distinct costimulatory domains mitigate HIV pathogenesis in vivo. Nat. Med. 2020, 26, 1776–1787. [Google Scholar] [CrossRef] [PubMed]
  201. Shimizu, S.; Ringpis, G.E.; Marsden, M.D.; Cortado, R.V.; Wilhalme, H.M.; Elashoff, D.; Zack, J.A.; Chen, I.S.; An, D.S. RNAi-Mediated CCR5 Knockdown Provides HIV-1 Resistance to Memory T Cells in Humanized BLT Mice. Mol. Ther. Nucleic Acids 2015, 4, e227. [Google Scholar] [CrossRef]
  202. Yi, G.; Choi, J.G.; Bharaj, P.; Abraham, S.; Dang, Y.; Kafri, T.; Alozie, O.; Manjunath, M.N.; Shankar, P. CCR5 Gene Editing of Resting CD4+ T Cells by Transient ZFN Expression From HIV Envelope Pseudotyped Nonintegrating Lentivirus Confers HIV-1 Resistance in Humanized Mice. Mol. Ther. Nucleic Acids 2014, 3, e198. [Google Scholar] [CrossRef]
  203. Anthony-Gonda, K.; Bardhi, A.; Ray, A.; Flerin, N.; Li, M.; Chen, W.; Ochsenbauer, C.; Kappes, J.C.; Krueger, W.; Worden, A.; et al. Multispecific anti-HIV duoCAR-T cells display broad in vitro antiviral activity and potent in vivo elimination of HIV-infected cells in a humanized mouse model. Sci. Transl. Med. 2019, 11, eaav5685. [Google Scholar] [CrossRef]
  204. Daharsh, L.; Zhang, J.; Ramer-Tait, A.; Li, Q. A Double Humanized BLT-mice Model Featuring a Stable Human-Like Gut Microbiome and Human Immune System. J. Vis. Exp. 2019, e59773. [Google Scholar] [CrossRef]
  205. Claiborne, D.T.; Dudek, T.E.; Maldini, C.R.; Power, K.A.; Ghebremichael, M.; Seung, E.; Mellors, E.F.; Vrbanac, V.D.; Krupp, K.; Bisesi, A.; et al. Immunization of BLT Humanized Mice Redirects T Cell Responses to Gag and Reduces Acute HIV-1 Viremia. J. Virol. 2019, 93, e00814-19. [Google Scholar] [CrossRef] [PubMed]
  206. Lavender, K.J.; Pace, C.; Sutter, K.; Messer, R.J.; Pouncey, D.L.; Cummins, N.W.; Natesampillai, S.; Zheng, J.; Goldsmith, J.; Widera, M.; et al. An advanced BLT-humanized mouse model for extended HIV-1 cure studies. AIDS 2018, 32, 1–10. [Google Scholar] [CrossRef]
  207. Marsden, M.D.; Zack, J.A. Humanized Mouse Models for Human Immunodeficiency Virus Infection. Annu. Rev. Virol. 2017, 4, 393–412. [Google Scholar] [CrossRef] [PubMed]
  208. Seung, E.; Dudek, T.E.; Allen, T.M.; Freeman, G.J.; Luster, A.D.; Tager, A.M. PD-1 blockade in chronically HIV-1-infected humanized mice suppresses viral loads. PLoS ONE 2013, 8, e77780. [Google Scholar] [CrossRef] [PubMed]
  209. Wahl, A.; Victor Garcia, J. The use of BLT humanized mice to investigate the immune reconstitution of the gastrointestinal tract. J. Immunol. Methods 2014, 410, 28–33. [Google Scholar] [CrossRef] [PubMed]
  210. Ringpis, G.E.; Shimizu, S.; Arokium, H.; Camba-Colon, J.; Carroll, M.V.; Cortado, R.; Xie, Y.; Kim, P.Y.; Sahakyan, A.; Lowe, E.L.; et al. Engineering HIV-1-resistant T-cells from short-hairpin RNA-expressing hematopoietic stem/progenitor cells in humanized BLT mice. PLoS ONE 2012, 7, e53492. [Google Scholar] [CrossRef]
  211. Vatakis, D.N.; Bristol, G.C.; Kim, S.G.; Levin, B.; Liu, W.; Radu, C.G.; Kitchen, S.G.; Zack, J.A. Using the BLT humanized mouse as a stem cell based gene therapy tumor model. J. Vis. Exp. 2012, e4181. [Google Scholar] [CrossRef]
  212. Denton, P.W.; Olesen, R.; Choudhary, S.K.; Archin, N.M.; Wahl, A.; Swanson, M.D.; Chateau, M.; Nochi, T.; Krisko, J.F.; Spagnuolo, R.A.; et al. Generation of HIV latency in humanized BLT mice. J. Virol. 2012, 86, 630–634. [Google Scholar] [CrossRef]
  213. Marsden, M.D.; Kovochich, M.; Suree, N.; Shimizu, S.; Mehta, R.; Cortado, R.; Bristol, G.; An, D.S.; Zack, J.A. HIV latency in the humanized BLT mouse. J. Virol. 2012, 86, 339–347. [Google Scholar] [CrossRef]
  214. Marsden, M.D.; Zhang, T.H.; Du, Y.; Dimapasoc, M.; Soliman, M.S.A.; Wu, X.; Kim, J.T.; Shimizu, A.; Schrier, A.; Wender, P.A.; et al. Tracking HIV Rebound following Latency Reversal Using Barcoded HIV. Cell Rep. Med. 2020, 1, 100162. [Google Scholar] [CrossRef] [PubMed]
  215. Yan, H.; Semple, K.M.; Gonzalez, C.M.; Howard, K.E. Bone marrow-liver-thymus (BLT) immune humanized mice as a model to predict cytokine release syndrome. Transl. Res. 2019, 210, 43–56. [Google Scholar] [CrossRef] [PubMed]
  216. Ginwala, R.; Caruso, B.; Khan, Z.K.; Pattekar, A.; Chew, G.M.; Corley, M.J.; Loonawat, R.; Jacobson, S.; Sreedhar, S.; Ndhlovu, L.C.; et al. HTLV-1 Infection and Neuropathogenesis in the Context of Rag1−/−γc−/− (RAG1-Hu) and BLT Mice. J. Neuroimmune Pharmacol. Off. J. Soc. NeuroImmune Pharmacol. 2017, 12, 504–520. [Google Scholar] [CrossRef] [PubMed]
  217. Crawford, L.B.; Tempel, R.; Streblow, D.N.; Kreklywich, C.; Smith, P.; Picker, L.J.; Nelson, J.A.; Caposio, P. Human Cytomegalovirus Induces Cellular and Humoral Virus-specific Immune Responses in Humanized BLT Mice. Sci. Rep. 2017, 7, 937. [Google Scholar] [CrossRef]
  218. Garcia-Beltran, W.F.; Claiborne, D.T.; Maldini, C.R.; Phelps, M.; Vrbanac, V.; Karpel, M.E.; Krupp, K.L.; Power, K.A.; Boutwell, C.L.; Balazs, A.B.; et al. Innate Immune Reconstitution in Humanized Bone Marrow-Liver-Thymus (HuBLT) Mice Governs Adaptive Cellular Immune Function and Responses to HIV-1 Infection. Front. Immunol. 2021, 12, 667393. [Google Scholar] [CrossRef]
  219. Kitsera, M.; Brunetti, J.E.; Rodriguez, E. Recent Developments in NSG and NRG Humanized Mouse Models for Their Use in Viral and Immune Research. Viruses 2023, 15, 478. [Google Scholar] [CrossRef]
  220. Lavender, K.J.; Pang, W.W.; Messer, R.J.; Duley, A.K.; Race, B.; Phillips, K.; Scott, D.; Peterson, K.E.; Chan, C.K.; Dittmer, U.; et al. BLT-humanized C57BL/6 Rag2−/−γc−/−CD47−/− mice are resistant to GVHD and develop B- and T-cell immunity to HIV infection. Blood 2013, 122, 4013–4020. [Google Scholar] [CrossRef] [PubMed]
  221. Rongvaux, A.; Willinger, T.; Martinek, J.; Strowig, T.; Gearty, S.V.; Teichmann, L.L.; Saito, Y.; Marches, F.; Halene, S.; Palucka, A.K.; et al. Development and function of human innate immune cells in a humanized mouse model. Nat. Biotechnol. 2014, 32, 364–372. [Google Scholar] [CrossRef] [PubMed]
  222. Coughlan, A.M.; Harmon, C.; Whelan, S.; O’Brien, E.C.; O’Reilly, V.P.; Crotty, P.; Kelly, P.; Ryan, M.; Hickey, F.B.; O’Farrelly, C.; et al. Myeloid Engraftment in Humanized Mice: Impact of Granulocyte-Colony Stimulating Factor Treatment and Transgenic Mouse Strain. Stem Cells Dev. 2016, 25, 530–541. [Google Scholar] [CrossRef]
  223. Billerbeck, E.; Barry, W.T.; Mu, K.; Dorner, M.; Rice, C.M.; Ploss, A. Development of human CD4+FoxP3+ regulatory T cells in human stem cell factor-, granulocyte-macrophage colony-stimulating factor-, and interleukin-3-expressing NOD-SCID IL2R γnull humanized mice. Blood 2011, 117, 3076–3086. [Google Scholar] [CrossRef]
  224. Wunderlich, M.; Chou, F.S.; Link, K.A.; Mizukawa, B.; Perry, R.L.; Carroll, M.; Mulloy, J.C. AML xenograft efficiency is significantly improved in NOD/SCID-IL2RG mice constitutively expressing human SCF, GM-CSF and IL-3. Leukemia 2010, 24, 1785–1788. [Google Scholar] [CrossRef]
  225. Zhang, J.; Lohani, S.C.; Cheng, Y.; Wang, T.; Guo, L.; Kim, W.K.; Gorantla, S.; Li, Q. Human Microglia Extensively Reconstitute in Humanized-BLT Mice With Human Interleukin-34 Transgene and Support HIV-1 Brain Infection. Front. Immunol. 2021, 12, 672415. [Google Scholar] [CrossRef]
  226. Abeynaike, S.A.; Huynh, T.R.; Mehmood, A.; Kim, T.; Frank, K.; Gao, K.; Zalfa, C.; Gandarilla, A.; Shultz, L.; Paust, S. Human Hematopoietic Stem Cell Engrafted IL-15 Transgenic NSG Mice Support Robust NK Cell Responses and Sustained HIV-1 Infection. Viruses 2023, 15, 365. [Google Scholar] [CrossRef] [PubMed]
  227. Shan, L.; Flavell, R.A.; Herndler-Brandstetter, D. Development of Humanized Mouse Models for Studying Human NK Cells in Health and Disease. Methods Mol. Biol. 2022, 2463, 53–66. [Google Scholar] [CrossRef]
  228. Patton, J.; Vuyyuru, R.; Siglin, A.; Root, M.; Manser, T. Evaluation of the efficiency of human immune system reconstitution in NSG mice and NSG mice containing a human HLA.A2 transgene using hematopoietic stem cells purified from different sources. J. Immunol. Methods 2015, 422, 13–21. [Google Scholar] [CrossRef]
  229. Danner, R.; Chaudhari, S.N.; Rosenberger, J.; Surls, J.; Richie, T.L.; Brumeanu, T.D.; Casares, S. Expression of HLA class II molecules in humanized NOD.Rag1KO.IL2RgcKO mice is critical for development and function of human T and B cells. PLoS ONE 2011, 6, e19826. [Google Scholar] [CrossRef] [PubMed]
  230. Cheng, L.; Yu, H.; Li, G.; Li, F.; Ma, J.; Li, J.; Chi, L.; Zhang, L.; Su, L. Type I interferons suppress viral replication but contribute to T cell depletion and dysfunction during chronic HIV-1 infection. JCI Insight 2017, 2, e94366. [Google Scholar] [CrossRef] [PubMed]
  231. McNab, F.; Mayer-Barber, K.; Sher, A.; Wack, A.; O’Garra, A. Type I interferons in infectious disease. Nat. Rev. Immunol. 2015, 15, 87–103. [Google Scholar] [CrossRef]
  232. Rotger, M.; Dalmau, J.; Rauch, A.; McLaren, P.; Bosinger, S.E.; Martinez, R.; Sandler, N.G.; Roque, A.; Liebner, J.; Battegay, M.; et al. Comparative transcriptomics of extreme phenotypes of human HIV-1 infection and SIV infection in sooty mangabey and rhesus macaque. J. Clin. Investig. 2011, 121, 2391–2400. [Google Scholar] [CrossRef]
  233. Mu, W.; Rezek, V.; Martin, H.; Carrillo, M.A.; Tomer, S.; Hamid, P.; Lizarraga, M.A.; Tibbe, T.D.; Yang, O.O.; Jamieson, B.D.; et al. Autophagy inducer rapamycin treatment reduces IFN-I-mediated Inflammation and improves anti-HIV-1 T cell response in vivo. JCI Insight 2022, 7, e159136. [Google Scholar] [CrossRef] [PubMed]
  234. Daskou, M.; Mu, W.; Sharma, M.; Vasilopoulos, H.; Heymans, R.; Ritou, E.; Rezek, V.; Hamid, P.; Kossyvakis, A.; Sen Roy, S.; et al. ApoA-I mimetics reduce systemic and gut inflammation in chronic treated HIV. PLoS Pathog. 2022, 18, e1010160. [Google Scholar] [CrossRef]
  235. Mu, W.; Sharma, M.; Heymans, R.; Ritou, E.; Rezek, V.; Hamid, P.; Kossyvakis, A.; Sen Roy, S.; Grijalva, V.; Chattopadhyay, A.; et al. Apolipoprotein A-I mimetics attenuate macrophage activation in chronic treated HIV. AIDS 2021, 35, 543–553. [Google Scholar] [CrossRef] [PubMed]
  236. Amand, M.; Adams, P.; Schober, R.; Iserentant, G.; Servais, J.Y.; Moutschen, M.; Seguin-Devaux, C. The anti-caspase 1 inhibitor VX-765 reduces immune activation, CD4+ T cell depletion, viral load, and total HIV-1 DNA in HIV-1 infected humanized mice. eLife 2023, 12, e83207. [Google Scholar] [CrossRef] [PubMed]
  237. McGary, C.S.; Silvestri, G.; Paiardini, M. Animal models for viral infection and cell exhaustion. Curr. Opin. HIV AIDS 2014, 9, 492–499. [Google Scholar] [CrossRef]
  238. Palmer, B.E.; Neff, C.P.; Lecureux, J.; Ehler, A.; Dsouza, M.; Remling-Mulder, L.; Korman, A.J.; Fontenot, A.P.; Akkina, R. In vivo blockade of the PD-1 receptor suppresses HIV-1 viral loads and improves CD4+ T cell levels in humanized mice. J. Immunol. 2013, 190, 211–219. [Google Scholar] [CrossRef] [PubMed]
  239. Proto, J.D.; Doran, A.C.; Subramanian, M.; Wang, H.; Zhang, M.; Sozen, E.; Rymond, C.C.; Kuriakose, G.; D’Agati, V.; Winchester, R.; et al. Hypercholesterolemia induces T cell expansion in humanized immune mice. J. Clin. Investig. 2018, 128, 2370–2375. [Google Scholar] [CrossRef]
  240. Day, E.A.; O’Neill, L.A.J. Targeting mitochondria to beat HIV-1. Nat. Immunol. 2021, 22, 398–399. [Google Scholar] [CrossRef]
  241. Castellano, P.; Prevedel, L.; Valdebenito, S.; Eugenin, E.A. HIV infection and latency induce a unique metabolic signature in human macrophages. Sci. Rep. 2019, 9, 3941. [Google Scholar] [CrossRef]
  242. Wahl, A.; Yao, W.; Liao, B.; Chateau, M.; Richardson, C.; Ling, L.; Franks, A.; Senthil, K.; Doyon, G.; Li, F.; et al. A germ-free humanized mouse model shows the contribution of resident microbiota to human-specific pathogen infection. Nat. Biotechnol. 2023. [Google Scholar] [CrossRef]
  243. Duncan, B.B.; Dunbar, C.E.; Ishii, K. Applying a clinical lens to animal models of CAR-T cell therapies. Mol. Ther. Methods Clin. Dev. 2022, 27, 17–31. [Google Scholar] [CrossRef]
  244. Cogels, M.M.; Rouas, R.; Ghanem, G.E.; Martinive, P.; Awada, A.; Van Gestel, D.; Krayem, M. Humanized Mice as a Valuable Pre-Clinical Model for Cancer Immunotherapy Research. Front. Oncol. 2021, 11, 784947. [Google Scholar] [CrossRef]
  245. Mu, W.; Carrillo, M.A.; Kitchen, S.G. Engineering CAR T Cells to Target the HIV Reservoir. Front. Cell. Infect. Microbiol. 2020, 10, 410. [Google Scholar] [CrossRef]
  246. Carrillo, M.A.; Zhen, A.; Kitchen, S.G. The Use of the Humanized Mouse Model in Gene Therapy and Immunotherapy for HIV and Cancer. Front. Immunol. 2018, 9, 746. [Google Scholar] [CrossRef] [PubMed]
  247. Hajduczki, A.; Danielson, D.T.; Elias, D.S.; Bundoc, V.; Scanlan, A.W.; Berger, E.A. A Trispecific Anti-HIV Chimeric Antigen Receptor Containing the CCR5 N-Terminal Region. Front. Cell. Infect. Microbiol. 2020, 10, 242. [Google Scholar] [CrossRef] [PubMed]
  248. Leibman, R.S.; Richardson, M.W.; Ellebrecht, C.T.; Maldini, C.R.; Glover, J.A.; Secreto, A.J.; Kulikovskaya, I.; Lacey, S.F.; Akkina, S.R.; Yi, Y.; et al. Supraphysiologic control over HIV-1 replication mediated by CD8 T cells expressing a re-engineered CD4-based chimeric antigen receptor. PLoS Pathog. 2017, 13, e1006613. [Google Scholar] [CrossRef] [PubMed]
  249. Liu, L.; Patel, B.; Ghanem, M.H.; Bundoc, V.; Zheng, Z.; Morgan, R.A.; Rosenberg, S.A.; Dey, B.; Berger, E.A. Novel CD4-Based Bispecific Chimeric Antigen Receptor Designed for Enhanced Anti-HIV Potency and Absence of HIV Entry Receptor Activity. J. Virol. 2015, 89, 6685–6694. [Google Scholar] [CrossRef] [PubMed]
  250. Imai, S.; Haga, S.; Kiyozuka, Y. Epitope characterization of MUC1 antibodies. Tumour Biol. 1998, 19 (Suppl. 1), 30–34. [Google Scholar] [CrossRef] [PubMed]
  251. Collins, A.; Morton, N.E. Likelihood ratios for DNA identification. Proc. Natl. Acad. Sci. USA 1994, 91, 6007–6011. [Google Scholar] [CrossRef]
  252. Marshall, B.E.; Soma, L.R.; Harp, J.R.; Neufeld, G.R.; Wurzel, H.A.; Dodd, D.C. Pulmonary function after exchange transfusion of stored blood in dogs. Ann. Surg. 1974, 179, 46–51. [Google Scholar] [CrossRef]
  253. Lim, R.M.; Rong, L.; Zhen, A.; Xie, J. A Universal CAR-NK Cell Targeting Various Epitopes of HIV-1 gp160. ACS Chem. Biol. 2020, 15, 2299–2310. [Google Scholar] [CrossRef]
  254. Ali, A.; Chiuppesi, F.; Nguyen, M.; Hausner, M.A.; Nguyen, J.; Kha, M.; Iniguez, A.; Wussow, F.; Diamond, D.J.; Yang, O.O. Chimeric Antigen Receptors Targeting Human Cytomegalovirus. J. Infect. Dis. 2020, 222, 853–862. [Google Scholar] [CrossRef] [PubMed]
  255. Hale, M.; Mesojednik, T.; Romano Ibarra, G.S.; Sahni, J.; Bernard, A.; Sommer, K.; Scharenberg, A.M.; Rawlings, D.J.; Wagner, T.A. Engineering HIV-Resistant, Anti-HIV Chimeric Antigen Receptor T Cells. Mol. Ther. J. Am. Soc. Gene Ther. 2017, 25, 570–579. [Google Scholar] [CrossRef] [PubMed]
  256. Zhen, A.; Carrillo, M.A.; Mu, W.; Rezek, V.; Martin, H.; Hamid, P.; Chen, I.S.Y.; Yang, O.O.; Zack, J.A.; Kitchen, S.G. Robust CAR-T memory formation and function via hematopoietic stem cell delivery. PLoS Pathog. 2021, 17, e1009404. [Google Scholar] [CrossRef] [PubMed]
  257. Zhen, A.; Peterson, C.W.; Carrillo, M.A.; Reddy, S.S.; Youn, C.S.; Lam, B.B.; Chang, N.Y.; Martin, H.A.; Rick, J.W.; Kim, J.; et al. Long-term persistence and function of hematopoietic stem cell-derived chimeric antigen receptor T cells in a nonhuman primate model of HIV/AIDS. PLoS Pathog. 2017, 13, e1006753. [Google Scholar] [CrossRef]
  258. Rust, B.J.; Kean, L.S.; Colonna, L.; Brandenstein, K.E.; Poole, N.H.; Obenza, W.; Enstrom, M.R.; Maldini, C.R.; Ellis, G.I.; Fennessey, C.M.; et al. Robust expansion of HIV CAR T cells following antigen boosting in ART-suppressed nonhuman primates. Blood 2020, 136, 1722–1734. [Google Scholar] [CrossRef]
  259. Jiang, Z.; Liang, H.; Pan, H.; Liang, Y.; Wang, H.; Yang, X.; Lu, P.; Zhang, X.; Yang, J.; Zhang, D.; et al. HIV-1-Specific CAR-T Cells With Cell-Intrinsic PD-1 Checkpoint Blockade Enhance Anti-HIV Efficacy in vivo. Front. Microbiol. 2021, 12, 684016. [Google Scholar] [CrossRef]
  260. Mylvaganam, G.; Yanez, A.G.; Maus, M.; Walker, B.D. Toward T Cell-Mediated Control or Elimination of HIV Reservoirs: Lessons From Cancer Immunology. Front. Immunol. 2019, 10, 2109. [Google Scholar] [CrossRef]
  261. Gumber, D.; Wang, L.D. Improving CAR-T immunotherapy: Overcoming the challenges of T cell exhaustion. eBioMedicine 2022, 77, 103941. [Google Scholar] [CrossRef] [PubMed]
  262. Kawalekar, O.U.; O’Connor, R.S.; Fraietta, J.A.; Guo, L.; McGettigan, S.E.; Posey, A.D., Jr.; Patel, P.R.; Guedan, S.; Scholler, J.; Keith, B.; et al. Distinct Signaling of Coreceptors Regulates Specific Metabolism Pathways and Impacts Memory Development in CAR T Cells. Immunity 2016, 44, 380–390. [Google Scholar] [CrossRef] [PubMed]
  263. Shen, L.; Xiao, Y.; Tian, J.; Lu, Z. Remodeling metabolic fitness: Strategies for improving the efficacy of chimeric antigen receptor T cell therapy. Cancer Lett. 2022, 529, 139–152. [Google Scholar] [CrossRef] [PubMed]
  264. Geltink, R.I.K.; Kyle, R.L.; Pearce, E.L. Unraveling the Complex Interplay Between T Cell Metabolism and Function. Annu. Rev. Immunol. 2018, 36, 461–488. [Google Scholar] [CrossRef]
  265. Phan, A.T.; Goldrath, A.W.; Glass, C.K. Metabolic and Epigenetic Coordination of T Cell and Macrophage Immunity. Immunity 2017, 46, 714–729. [Google Scholar] [CrossRef]
  266. Li, Y.; Tang, J.; Jiang, J.; Chen, Z. Metabolic checkpoints and novel approaches for immunotherapy against cancer. Int. J. Cancer 2022, 150, 195–207. [Google Scholar] [CrossRef]
  267. Zimmermannova, O.; Caiado, I.; Ferreira, A.G.; Pereira, C.F. Cell Fate Reprogramming in the Era of Cancer Immunotherapy. Front. Immunol. 2021, 12, 714822. [Google Scholar] [CrossRef]
  268. Rangel Rivera, G.O.; Knochelmann, H.M.; Dwyer, C.J.; Smith, A.S.; Wyatt, M.M.; Rivera-Reyes, A.M.; Thaxton, J.E.; Paulos, C.M. Fundamentals of T Cell Metabolism and Strategies to Enhance Cancer Immunotherapy. Front. Immunol. 2021, 12, 645242. [Google Scholar] [CrossRef]
  269. Jaccard, A.; Wyss, T.; Maldonado-Perez, N.; Rath, J.A.; Bevilacqua, A.; Peng, J.J.; Lepez, A.; Von Gunten, C.; Franco, F.; Kao, K.C.; et al. Reductive carboxylation epigenetically instructs T cell differentiation. Nature 2023, 621, 849–856. [Google Scholar] [CrossRef]
Figure 1. Chronic HIV infection leads to T cells metabolic stress, immune activation and T cell dysfunction. Despite ART, HIV infection induces persistent immune activation and metabolic alterations in T cells, marked by increased Type I IFN, heightened Reactive Oxygen Species (ROS), mitochondrial dysfunction, etc. Both persistent immune activation and metabolic stress eventually contribute to T cell exhaustion.
Figure 1. Chronic HIV infection leads to T cells metabolic stress, immune activation and T cell dysfunction. Despite ART, HIV infection induces persistent immune activation and metabolic alterations in T cells, marked by increased Type I IFN, heightened Reactive Oxygen Species (ROS), mitochondrial dysfunction, etc. Both persistent immune activation and metabolic stress eventually contribute to T cell exhaustion.
Viruses 16 00219 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Mu, W.; Patankar, V.; Kitchen, S.; Zhen, A. Examining Chronic Inflammation, Immune Metabolism, and T Cell Dysfunction in HIV Infection. Viruses 2024, 16, 219. https://doi.org/10.3390/v16020219

AMA Style

Mu W, Patankar V, Kitchen S, Zhen A. Examining Chronic Inflammation, Immune Metabolism, and T Cell Dysfunction in HIV Infection. Viruses. 2024; 16(2):219. https://doi.org/10.3390/v16020219

Chicago/Turabian Style

Mu, Wenli, Vaibhavi Patankar, Scott Kitchen, and Anjie Zhen. 2024. "Examining Chronic Inflammation, Immune Metabolism, and T Cell Dysfunction in HIV Infection" Viruses 16, no. 2: 219. https://doi.org/10.3390/v16020219

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop